1
|
Szarka G, Ganczer A, Balogh M, Tengölics ÁJ, Futácsi A, Kenyon G, Pan F, Kovács-Öller T, Völgyi B. Gap junctions fine-tune ganglion cell signals to equalize response kinetics within a given electrically coupled array. iScience 2024; 27:110099. [PMID: 38947503 PMCID: PMC11214328 DOI: 10.1016/j.isci.2024.110099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/06/2024] [Accepted: 05/22/2024] [Indexed: 07/02/2024] Open
Abstract
Retinal ganglion cells (RGCs) summate inputs and forward a spike train code to the brain in the form of either maintained spiking (sustained) or a quickly decaying brief spike burst (transient). We report diverse response transience values across the RGC population and, contrary to the conventional transient/sustained scheme, responses with intermediary characteristics are the most abundant. Pharmacological tests showed that besides GABAergic inhibition, gap junction (GJ)-mediated excitation also plays a pivotal role in shaping response transience and thus visual coding. More precisely GJs connecting RGCs to nearby amacrine and RGCs play a defining role in the process. These GJs equalize kinetic features, including the response transience of transient OFF alpha (tOFFα) RGCs across a coupled array. We propose that GJs in other coupled neuron ensembles in the brain are also critical in the harmonization of response kinetics to enhance the population code and suit a corresponding task.
Collapse
Affiliation(s)
- Gergely Szarka
- University of Pécs, Szentágothai Research Centre, Pécs, Hungary
- University of Pécs, Department of Neurobiology, Pécs, Hungary
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, Pécs, Hungary
- Center for Neuroscience, University of Pécs, Pécs, Hungary
- SzKK Imaging Core Facility, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Alma Ganczer
- University of Pécs, Szentágothai Research Centre, Pécs, Hungary
- University of Pécs, Department of Neurobiology, Pécs, Hungary
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, Pécs, Hungary
- Center for Neuroscience, University of Pécs, Pécs, Hungary
| | - Márton Balogh
- University of Pécs, Szentágothai Research Centre, Pécs, Hungary
- University of Pécs, Department of Neurobiology, Pécs, Hungary
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, Pécs, Hungary
- Center for Neuroscience, University of Pécs, Pécs, Hungary
| | - Ádám Jonatán Tengölics
- University of Pécs, Szentágothai Research Centre, Pécs, Hungary
- University of Pécs, Department of Neurobiology, Pécs, Hungary
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, Pécs, Hungary
- Center for Neuroscience, University of Pécs, Pécs, Hungary
| | - Anett Futácsi
- University of Pécs, Szentágothai Research Centre, Pécs, Hungary
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, Pécs, Hungary
- Center for Neuroscience, University of Pécs, Pécs, Hungary
- SzKK Imaging Core Facility, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | | | - Feng Pan
- The Hong Kong Polytechnic University, Hong Kong, China
| | - Tamás Kovács-Öller
- University of Pécs, Szentágothai Research Centre, Pécs, Hungary
- University of Pécs, Department of Neurobiology, Pécs, Hungary
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, Pécs, Hungary
- Center for Neuroscience, University of Pécs, Pécs, Hungary
- SzKK Imaging Core Facility, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Béla Völgyi
- University of Pécs, Szentágothai Research Centre, Pécs, Hungary
- University of Pécs, Department of Neurobiology, Pécs, Hungary
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, Pécs, Hungary
- Center for Neuroscience, University of Pécs, Pécs, Hungary
| |
Collapse
|
2
|
Ribelayga CP, O’Brien J. When microscopy and electrophysiology meet connectomics-Steve Massey's contribution to unraveling the structure and function of the rod/cone gap junction. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1305131. [PMID: 38983007 PMCID: PMC11182179 DOI: 10.3389/fopht.2023.1305131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 10/31/2023] [Indexed: 07/11/2024]
Abstract
Electrical synapses, formed of gap junctions, are ubiquitous components of the central nervous system (CNS) that shape neuronal circuit connectivity and dynamics. In the retina, electrical synapses can create a circuit, control the signal-to-noise ratio in individual neurons, and support the coordinated neuronal firing of ganglion cells, hence, regulating signal processing at the network, single-cell, and dendritic level. We, the authors, and Steve Massey have had a long interest in gap junctions in retinal circuits, in general, and in the network of photoreceptors, in particular. Our combined efforts, based on a wide array of techniques of molecular biology, microscopy, and electrophysiology, have provided fundamental insights into the molecular structure and properties of the rod/cone gap junction. Yet, a full understanding of how rod/cone coupling controls circuit dynamics necessitates knowing its operating range. It is well established that rod/cone coupling can be greatly reduced or eliminated by bright-light adaptation or pharmacological treatment; however, the upper end of its dynamic range has long remained elusive. This held true until Steve Massey's recent interest for connectomics led to the development of a new strategy to assess this issue. The effort proved effective in establishing, with precision, the connectivity rules between rods and cones and estimating the theoretical upper limit of rod/cone electrical coupling. Comparing electrophysiological measurements and morphological data indicates that under pharmacological manipulation, rod/cone coupling can reach the theoretical maximum of its operating range, implying that, under these conditions, all the gap junction channels present at the junctions are open. As such, channel open probability is likely the main determinant of rod/cone coupling that can change momentarily in a time-of-day- and light-dependent manner. In this article we briefly review our current knowledge of the molecular structure of the rod/cone gap junction and of the mechanisms behind its modulation, and we highlight the recent work led by Steve Massey. Steve's contribution has been critical toward asserting the modulation depth of rod/cone coupling as well as elevating the rod/cone gap junction as one of the most suitable models to examine the role of electrical synapses and their plasticity in neural processing.
Collapse
Affiliation(s)
- Christophe P. Ribelayga
- Department of Vision Sciences, University of Houston College of Optometry, Houston, TX, United States
| | | |
Collapse
|
3
|
Maruyama T, Ishii T, Kaneda M. Starburst amacrine cells form gap junctions in the early postnatal stage of the mouse retina. Front Cell Neurosci 2023; 17:1173579. [PMID: 37293630 PMCID: PMC10244514 DOI: 10.3389/fncel.2023.1173579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/03/2023] [Indexed: 06/10/2023] Open
Abstract
Although gap junctional coupling in the developing retina is important for the maturation of neuronal networks, its role in the development of individual neurons remains unclear. Therefore, we herein investigated whether gap junctional coupling by starburst amacrine cells (SACs), a key neuron for the formation of direction selectivity, occurs during the developmental stage in the mouse retina. Neurobiotin-injected SACs coupled with many neighboring cells before eye-opening. The majority of tracer-coupled cells were retinal ganglion cells, and tracer coupling was not detected between SACs. The number of tracer-coupled cells significantly decreased after eye-opening and mostly disappeared by postnatal day 28 (P28). Membrane capacitance (Cm), an indicator of the formation of electrical coupling with gap junctions, was larger in SACs before than after eye-opening. The application of meclofenamic acid, a gap junction blocker, reduced the Cm of SACs. Gap junctional coupling by SACs was regulated by dopamine D1 receptors before eye-opening. In contrast, the reduction in gap junctional coupling after eye-opening was not affected by visual experience. At the mRNA level, 4 subtypes of connexins (23, 36, 43, and 45) were detected in SACs before eye-opening. Connexin 43 expression levels significantly decreased after eye-opening. These results indicate that gap junctional coupling by SACs occurs during the developmental period and suggest that the elimination of gap junctions proceeds with the innate system.
Collapse
|
4
|
Esteban-Linares A, Zhang X, Lee HH, Risner ML, Weiss SM, Xu YQ, Levine E, Li D. Graphene-based microfluidic perforated microelectrode arrays for retinal electrophysiological studies. LAB ON A CHIP 2023; 23:2193-2205. [PMID: 36891773 PMCID: PMC10159897 DOI: 10.1039/d3lc00064h] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Perforated microelectrode arrays (pMEAs) have become essential tools for ex vivo retinal electrophysiological studies. pMEAs increase the nutrient supply to the explant and alleviate the accentuated curvature of the retina, allowing for long-term culture and intimate contacts between the retina and electrodes for electrophysiological measurements. However, commercial pMEAs are not compatible with in situ high-resolution optical imaging and lack the capability of controlling the local microenvironment, which are highly desirable features for relating function to anatomy and probing physiological and pathological mechanisms in retina. Here we report on microfluidic pMEAs (μpMEAs) that combine transparent graphene electrodes and the capability of locally delivering chemical stimulation. We demonstrate the potential of μpMEAs by measuring the electrical response of ganglion cells to locally delivered high K+ stimulation under controlled microenvironments. Importantly, the capability for high-resolution confocal imaging of the retina tissue on top of the graphene electrodes allows for further analyses of the electrical signal source. The new capabilities provided by μpMEAs could allow for retinal electrophysiology assays to address key questions in retinal circuitry studies.
Collapse
Affiliation(s)
| | - Xiaosi Zhang
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Hannah H Lee
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| | - Michael L Risner
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| | - Sharon M Weiss
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Department of Physics and Astronomy, Vanderbilt University, Nashville, TN, 37235, USA
| | - Ya-Qiong Xu
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Department of Physics and Astronomy, Vanderbilt University, Nashville, TN, 37235, USA
| | - Edward Levine
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37235, USA
| | - Deyu Li
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN, 37235, USA.
| |
Collapse
|
5
|
Urbán P, Pöstyéni E, Czuni L, Herczeg R, Fekete C, Gábriel R, Kovács-Valasek A. miRNA Profiling of Developing Rat Retina in the First Three Postnatal Weeks. Cell Mol Neurobiol 2023:10.1007/s10571-023-01347-3. [PMID: 37084144 DOI: 10.1007/s10571-023-01347-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 04/04/2023] [Indexed: 04/22/2023]
Abstract
The morphogenesis of the mammalian retina depends on the precise control of gene expression during development. Small non-coding RNAs, including microRNAs play profound roles in various physiological and pathological processes via gene expression regulation. A systematic analysis of the expression profile of small non-coding RNAs in developing Wistar rat retinas (postnatally day 5 (P5), P7, P10, P15 and P21) was executed using IonTorrent PGM next-generation sequencing technique to reveal the crucial players in the early postnatal retinogenesis. Our analysis reveals extensive regulatory potential of microRNAs during retinal development. We found a group of microRNAs that show constant high abundance (miR-19, miR-101; miR-181, miR-183, miR-124 and let-7) during the development process. Others are present only in the early stages (miR-20a, miR-206, miR-133, miR-466, miR-1247, miR-3582), or at later stages (miR-29, miR-96, miR-125, miR-344 or miR-664). Further miRNAs were detected which are differentially expressed in time. Finally, pathway enrichment analysis has revealed 850 predicted target genes that mainly participate in lipid-, amino acid- and glycan metabolisms in the examined time-period (P5-P21). P5-P7 transition revealed the importance of miRNAs in glutamatergic synapse and gap junction pathways. Significantly downregulated miRNAs rno-miR-30c1 and 2, rno-miR-205 and rno-miR-503 were detected to target Prkx (ENSRNOG00000003696), Adcy6 (ENSRNOG00000011587), Gnai3 (ENSRNOG00000019465) and Gja1 (ENSRNOG00000000805) genes. The dataset described here will be a valuable resource for clarifying new regulatory mechanisms for retinal development and will greatly contribute to our understanding of the divergence and function of microRNAs.
Collapse
Affiliation(s)
- Péter Urbán
- János Szentágothai Research Centre, University of Pécs, Pecs, Hungary
- Department of General and Environmental Microbiology, University of Pécs, Pecs, Hungary
| | - Etelka Pöstyéni
- Experimental Zoology and Neurobiology, University of Pécs, Pecs, Hungary
| | - Lilla Czuni
- János Szentágothai Research Centre, University of Pécs, Pecs, Hungary
| | - Róbert Herczeg
- János Szentágothai Research Centre, University of Pécs, Pecs, Hungary
| | - Csaba Fekete
- Department of General and Environmental Microbiology, University of Pécs, Pecs, Hungary
| | - Róbert Gábriel
- János Szentágothai Research Centre, University of Pécs, Pecs, Hungary
- Experimental Zoology and Neurobiology, University of Pécs, Pecs, Hungary
| | | |
Collapse
|
6
|
Szarka G, Hoffmann G, Kovács-Öller T, Völgyi B. Serotonin is a gap junction-permeable neuronal tracer in the mouse retina. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1151024. [PMID: 38983061 PMCID: PMC11182087 DOI: 10.3389/fopht.2023.1151024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 02/27/2023] [Indexed: 07/11/2024]
Abstract
Introduction Gap junctions are dynamically modulated bridges allowing the transcellular passage of ions and small molecules with a molecular mass of up to 1 kDa, a mechanism utilized for molecular communication purposes by living cells. This same mechanism is also exploited by scientists to reveal the existence of gap junction contacts by the cell-to-cell movement of tracers. However, multiple labeling experiments require the availability of multiple gap junction-permeable tracers. Methods To this end, we utilized the well-known transient OFF alpha retinal ganglion cell (RGC)-coupled array as a model system to study and compare the transjunctional movement of neurobiotin (NB), a commonly used tracer, and serotonin, a recently identified tracer. Results Although the transjunctional movement of serotonin has been established in cell cultures, here we show, for the first time, that serotonin is also a potent tracer in in vitro tissue. In addition, serotonin is lighter than the classical gap junction-permeable NB, and thus, we expected that tracer movement would be comparable to or better than that of serotonin. We found that intracellular serotonin injections result in the labeling of the coupled transient OFF alpha RGC array very similar to those of the classical NB-labeled arrays. Both serotonin and NB-injected transient OFF alpha RGCs displayed the well-known pattern with coupled RGCs and a cohort of coupled wide-field amacrine cells (ACs). Discussion By using morphological characteristics, we confirm that the serotonin and the NB-coupled AC arrays are identical, and thereby confirm that serotonin is a potent gap junction-permeable tracer and can be readily used as an alternative to NB in in vitro tissue. Moreover, serotonin can be utilized in parallel with other dyes or tracers, enabling the use of multiple labels in the same material.
Collapse
Affiliation(s)
- Gergely Szarka
- Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Comparative Anatomy and Developmental Biology, University of Pécs, Pécs, Hungary
- Center for Neuroscience, University of Pécs, Pécs, Hungary
- NEURON-066 Rethealthsi Research Group, Pécs, Hungary
| | - Gyula Hoffmann
- Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Comparative Anatomy and Developmental Biology, University of Pécs, Pécs, Hungary
- Center for Neuroscience, University of Pécs, Pécs, Hungary
| | - Tamás Kovács-Öller
- Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Comparative Anatomy and Developmental Biology, University of Pécs, Pécs, Hungary
- Center for Neuroscience, University of Pécs, Pécs, Hungary
- NEURON-066 Rethealthsi Research Group, Pécs, Hungary
| | - Béla Völgyi
- Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Comparative Anatomy and Developmental Biology, University of Pécs, Pécs, Hungary
- Center for Neuroscience, University of Pécs, Pécs, Hungary
- NEURON-066 Rethealthsi Research Group, Pécs, Hungary
| |
Collapse
|
7
|
Cha S, Ahn J, Jeong Y, Lee YH, Kim HK, Lee D, Yoo Y, Goo YS. Stage-Dependent Changes of Visual Function and Electrical Response of the Retina in the rd10 Mouse Model. Front Cell Neurosci 2022; 16:926096. [PMID: 35936494 PMCID: PMC9345760 DOI: 10.3389/fncel.2022.926096] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/23/2022] [Indexed: 12/28/2022] Open
Abstract
One of the critical prerequisites for the successful development of retinal prostheses is understanding the physiological features of retinal ganglion cells (RGCs) in the different stages of retinal degeneration (RD). This study used our custom-made rd10 mice, C57BL/6-Pde6bem1(R560C)Dkl/Korl mutated on the Pde6b gene in C57BL/6J mouse with the CRISPR/Cas9-based gene-editing method. We selected the postnatal day (P) 45, P70, P140, and P238 as representative ages for RD stages. The optomotor response measured the visual acuity across degeneration stages. At P45, the rd10 mice exhibited lower visual acuity than wild-type (WT) mice. At P140 and older, no optomotor response was observed. We classified RGC responses to the flashed light into ON, OFF, and ON/OFF RGCs via in vitro multichannel recording. With degeneration, the number of RGCs responding to the light stimulation decreased in all three types of RGCs. The OFF response disappeared faster than the ON response with older postnatal ages. We elicited RGC spikes with electrical stimulation and analyzed the network-mediated RGC response in the rd10 mice. Across all postnatal ages, the spikes of rd10 RGCs were less elicited by pulse amplitude modulation than in WT RGCs. The ratio of RGCs showing multiple peaks of spike burst increased in older ages. The electrically evoked RGC spikes by the pulse amplitude modulation differ across postnatal ages. Therefore, degeneration stage-dependent stimulation strategies should be considered for developing retinal prosthesis and successful vision restoration.
Collapse
Affiliation(s)
- Seongkwang Cha
- Department of Physiology, Chungbuk National University School of Medicine, Cheongju, South Korea
| | - Jungryul Ahn
- Department of Physiology, Chungbuk National University School of Medicine, Cheongju, South Korea
| | - Yurim Jeong
- Department of Physiology, Chungbuk National University School of Medicine, Cheongju, South Korea
| | - Yong Hee Lee
- Department of Biochemistry, Chungbuk National University School of Medicine, Cheongju, South Korea
| | - Hyong Kyu Kim
- Department of Microbiology, Chungbuk National University School of Medicine, Cheongju, South Korea
| | - Daekee Lee
- Department of Life Science, Ewha Womans University, Seoul, South Korea
| | - Yongseok Yoo
- Department of Electronics Engineering, Incheon National University, Incheon, South Korea
- *Correspondence: Yongseok Yoo,
| | - Yong Sook Goo
- Department of Physiology, Chungbuk National University School of Medicine, Cheongju, South Korea
- Yong Sook Goo,
| |
Collapse
|
8
|
Szarka G, Balogh M, Tengölics ÁJ, Ganczer A, Völgyi B, Kovács-Öller T. The role of gap junctions in cell death and neuromodulation in the retina. Neural Regen Res 2021; 16:1911-1920. [PMID: 33642359 PMCID: PMC8343308 DOI: 10.4103/1673-5374.308069] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/14/2020] [Accepted: 01/11/2021] [Indexed: 12/26/2022] Open
Abstract
Vision altering diseases, such as glaucoma, diabetic retinopathy, age-related macular degeneration, myopia, retinal vascular disease, traumatic brain injuries and others cripple many lives and are projected to continue to cause anguish in the foreseeable future. Gap junctions serve as an emerging target for neuromodulation and possible regeneration as they directly connect healthy and/or diseased cells, thereby playing a crucial role in pathophysiology. Since they are permeable for macromolecules, able to cross the cellular barriers, they show duality in illness as a cause and as a therapeutic target. In this review, we take recent advancements in gap junction neuromodulation (pharmacological blockade, gene therapy, electrical and light stimulation) into account, to show the gap junction's role in neuronal cell death and the possible routes of rescuing neuronal and glial cells in the retina succeeding illness or injury.
Collapse
Affiliation(s)
- Gergely Szarka
- János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, Budapest, Hungary
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| | - Márton Balogh
- János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, Budapest, Hungary
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| | - Ádám J. Tengölics
- János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, Budapest, Hungary
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| | - Alma Ganczer
- János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, Budapest, Hungary
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| | - Béla Völgyi
- János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, Budapest, Hungary
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
- Medical School, University of Pécs, Pécs, Hungary
| | - Tamás Kovács-Öller
- János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, Budapest, Hungary
- Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|
9
|
Fusz K, Kovács-Öller T, Kóbor P, Szabó-Meleg E, Völgyi B, Buzás P, Telkes I. Regional Variation of Gap Junctional Connections in the Mammalian Inner Retina. Cells 2021; 10:2396. [PMID: 34572046 PMCID: PMC8466939 DOI: 10.3390/cells10092396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/04/2021] [Accepted: 09/07/2021] [Indexed: 11/24/2022] Open
Abstract
The retinas of many species show regional specialisations that are evident in the differences in the processing of visual input from different parts of the visual field. Regional specialisation is thought to reflect an adaptation to the natural visual environment, optical constraints, and lifestyle of the species. Yet, little is known about regional differences in synaptic circuitry. Here, we were interested in the topographical distribution of connexin-36 (Cx36), the major constituent of electrical synapses in the retina. We compared the retinas of mice, rats, and cats to include species with different patterns of regional specialisations in the analysis. First, we used the density of Prox1-immunoreactive amacrine cells as a marker of any regional specialisation, with higher cell density signifying more central regions. Double-labelling experiments showed that Prox1 is expressed in AII amacrine cells in all three species. Interestingly, large Cx36 plaques were attached to about 8-10% of Prox1-positive amacrine cell somata, suggesting the strong electrical coupling of pairs or small clusters of cell bodies. When analysing the regional changes in the volumetric density of Cx36-immunoreactive plaques, we found a tight correlation with the density of Prox1-expressing amacrine cells in the ON, but not in the OFF sublamina in all three species. The results suggest that the relative contribution of electrical synapses to the ON- and OFF-pathways of the retina changes with retinal location, which may contribute to functional ON/OFF asymmetries across the visual field.
Collapse
Affiliation(s)
- Katalin Fusz
- Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary; (K.F.); (P.K.); (I.T.)
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Centre for Neuroscience, University of Pécs, 7624 Pécs, Hungary
| | - Tamás Kovács-Öller
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Centre for Neuroscience, University of Pécs, 7624 Pécs, Hungary
- MTA-PTE NAP-2 Retinal Electrical Synapses Research Group, 7624 Pécs, Hungary
| | - Péter Kóbor
- Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary; (K.F.); (P.K.); (I.T.)
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Centre for Neuroscience, University of Pécs, 7624 Pécs, Hungary
| | - Edina Szabó-Meleg
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Institute of Biophysics, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Béla Völgyi
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Centre for Neuroscience, University of Pécs, 7624 Pécs, Hungary
- MTA-PTE NAP-2 Retinal Electrical Synapses Research Group, 7624 Pécs, Hungary
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary
| | - Péter Buzás
- Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary; (K.F.); (P.K.); (I.T.)
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Centre for Neuroscience, University of Pécs, 7624 Pécs, Hungary
| | - Ildikó Telkes
- Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary; (K.F.); (P.K.); (I.T.)
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Centre for Neuroscience, University of Pécs, 7624 Pécs, Hungary
| |
Collapse
|
10
|
Zhi Z, Xiang J, Fu Q, Pei X, Zhou D, Cao Y, Xie L, Zhang S, Chen S, Qu J, Zhou X. The Role of Retinal Connexins Cx36 and Horizontal Cell Coupling in Emmetropization in Guinea Pigs. Invest Ophthalmol Vis Sci 2021; 62:27. [PMID: 34283211 PMCID: PMC8300059 DOI: 10.1167/iovs.62.9.27] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 06/24/2021] [Indexed: 11/24/2022] Open
Abstract
Purpose The purpose of this study was to determine whether retinal gap junctions (GJs) via connexin 36 (Cx36, mediating coupling of many retinal cell types) and horizontal cell (HC-HC) coupling, are involved in emmetropization. Methods Guinea pigs (3 weeks old) were monocularly form deprived (FD) or raised without FD (in normal visual [NV] environment) for 2 days or 4 weeks; alternatively, they wore a -4 D lens (hyperopic defocus [HD]) or 0 D lens for 2 days or 1 week. FD and NV eyes received daily subconjunctival injections of a nonspecific GJ-uncoupling agent, 18-β-Glycyrrhetinic Acid (18-β-GA). The amounts of total Cx36 and of phosphorylated Cx36 (P-Cx36; activated state that increases cell-cell coupling), in the inner and outer plexiform layers (IPLs and OPLs), were evaluated by quantitative immunofluorescence (IF), and HC-HC coupling was evaluated by cut-loading with neurobiotin. Results FD per se (excluding effect of light-attenuation) increased HC-HC coupling in OPL, whereas HD did not affect it. HD for 2 days or 1 week had no significant effect on retinal content of Cx36 or P-Cx36. FD for 4 weeks decreased the total amounts of Cx36 and P-Cx36, and the P-Cx36/Cx36 ratio, in the IPL. Subconjunctival 18-β-GA induced myopia in NV eyes and increased the myopic shifts in FD eyes, while reducing the amounts of Cx36 and P-Cx36 in both the IPL and OPL. Conclusions These results suggest that cell-cell coupling via GJs containing Cx36 (particularly those in the IPL) plays a role in emmetropization and form deprivation myopia (FDM) in mammals. Although both FD and 18-β-GA induced myopia, they had opposite effects on HC-HC coupling. These findings suggest that HC-HC coupling in the OPL might not play a significant role in emmetropization and myopia development.
Collapse
Affiliation(s)
- Zhina Zhi
- School of Optometry and Ophthalmology, and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health People's Republic of China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China
| | - Jing Xiang
- School of Optometry and Ophthalmology, and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health People's Republic of China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China
| | - Qian Fu
- School of Optometry and Ophthalmology, and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health People's Republic of China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China
| | - Xiaomeng Pei
- School of Optometry and Ophthalmology, and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health People's Republic of China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China
| | - Dengke Zhou
- School of Optometry and Ophthalmology, and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health People's Republic of China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China
| | - Yuqing Cao
- School of Optometry and Ophthalmology, and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health People's Republic of China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China
| | - Liqin Xie
- School of Optometry and Ophthalmology, and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health People's Republic of China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China
| | - Sen Zhang
- School of Optometry and Ophthalmology, and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health People's Republic of China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China
| | - Si Chen
- School of Optometry and Ophthalmology, and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health People's Republic of China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China
| | - Jia Qu
- School of Optometry and Ophthalmology, and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health People's Republic of China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China
| | - Xiangtian Zhou
- School of Optometry and Ophthalmology, and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health People's Republic of China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China
| |
Collapse
|
11
|
Chen AM, Azar SS, Harris A, Brecha NC, Pérez de Sevilla Müller L. PTEN Expression Regulates Gap Junction Connectivity in the Retina. Front Neuroanat 2021; 15:629244. [PMID: 34093139 PMCID: PMC8172595 DOI: 10.3389/fnana.2021.629244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/26/2021] [Indexed: 12/16/2022] Open
Abstract
Manipulation of the phosphatase and tensin homolog (PTEN) pathway has been suggested as a therapeutic approach to treat or prevent vision loss due to retinal disease. In this study, we investigated the effects of deleting one copy of Pten in a well-characterized class of retinal ganglion cells called α-ganglion cells in the mouse retina. In Pten +/- retinas, α-ganglion cells did not exhibit major changes in their dendritic structure, although most cells developed a few, unusual loop-forming dendrites. By contrast, α-ganglion cells exhibited a significant decrease in heterologous and homologous gap junction mediated cell coupling with other retinal ganglion and amacrine cells. Additionally, the majority of OFF α-ganglion cells (12/18 cells) formed novel coupling to displaced amacrine cells. The number of connexin36 puncta, the predominant connexin that mediates gap junction communication at electrical synapses, was decreased by at least 50% on OFF α-ganglion cells. Reduced and incorrect gap junction connectivity of α-ganglion cells will affect their functional properties and alter visual image processing in the retina. The anomalous connectivity of retinal ganglion cells would potentially limit future therapeutic approaches involving manipulation of the Pten pathway for treating ganglion cell degeneration in diseases like glaucoma, traumatic brain injury, Parkinson's, and Alzheimer's diseases.
Collapse
Affiliation(s)
- Ashley M. Chen
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California, Los Angeles, Los Angeles, CA, United States
| | - Shaghauyegh S. Azar
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California, Los Angeles, Los Angeles, CA, United States
| | - Alexander Harris
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California, Los Angeles, Los Angeles, CA, United States
| | - Nicholas C. Brecha
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California, Los Angeles, Los Angeles, CA, United States
- Stein Eye Institute, David Geffen School of Medicine at Los Angeles, University of California, Los Angeles, Los Angeles, CA, United States
- CURE Digestive Diseases Research Center, David Geffen School of Medicine at Los Angeles, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Administration Greater Los Angeles Health System, Los Angeles, CA, United States
| | - Luis Pérez de Sevilla Müller
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
12
|
Cangiano L, Asteriti S. Interphotoreceptor coupling: an evolutionary perspective. Pflugers Arch 2021; 473:1539-1554. [PMID: 33988778 PMCID: PMC8370920 DOI: 10.1007/s00424-021-02572-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 04/13/2021] [Accepted: 04/23/2021] [Indexed: 12/16/2022]
Abstract
In the vertebrate retina, signals generated by cones of different spectral preference and by highly sensitive rod photoreceptors interact at various levels to extract salient visual information. The first opportunity for such interaction is offered by electrical coupling of the photoreceptors themselves, which is mediated by gap junctions located at the contact points of specialised cellular processes: synaptic terminals, telodendria and radial fins. Here, we examine the evolutionary pressures for and against interphotoreceptor coupling, which are likely to have shaped how coupling is deployed in different species. The impact of coupling on signal to noise ratio, spatial acuity, contrast sensitivity, absolute and increment threshold, retinal signal flow and colour discrimination is discussed while emphasising available data from a variety of vertebrate models spanning from lampreys to primates. We highlight the many gaps in our knowledge, persisting discrepancies in the literature, as well as some major unanswered questions on the actual extent and physiological role of cone-cone, rod-cone and rod-rod communication. Lastly, we point toward limited but intriguing evidence suggestive of the ancestral form of coupling among ciliary photoreceptors.
Collapse
Affiliation(s)
- Lorenzo Cangiano
- Dept. of Translational Research, University of Pisa, Via San Zeno 31, 56123, Pisa, Italy.
| | - Sabrina Asteriti
- Dept. of Translational Research, University of Pisa, Via San Zeno 31, 56123, Pisa, Italy
| |
Collapse
|
13
|
Choi BJ, Chen YCD, Desplan C. Building a circuit through correlated spontaneous neuronal activity in the developing vertebrate and invertebrate visual systems. Genes Dev 2021; 35:677-691. [PMID: 33888564 PMCID: PMC8091978 DOI: 10.1101/gad.348241.121] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
During the development of the vertebrate nervous systems, genetic programs assemble an immature circuit that is subsequently refined by neuronal activity evoked by external stimuli. However, prior to sensory experience, the intrinsic property of the developing nervous system also triggers correlated network-level neuronal activity, with retinal waves in the developing vertebrate retina being the best documented example. Spontaneous activity has also been found in the visual system of Drosophila Here, we compare the spontaneous activity of the developing visual system between mammalian and Drosophila and suggest that Drosophila is an emerging model for mechanistic and functional studies of correlated spontaneous activity.
Collapse
Affiliation(s)
- Ben Jiwon Choi
- Department of Biology, New York University, New York, New York 10003, USA
| | | | - Claude Desplan
- Department of Biology, New York University, New York, New York 10003, USA
| |
Collapse
|
14
|
Fournel R, Hartveit E, Veruki ML. Differential Contribution of Gap Junctions to the Membrane Properties of ON- and OFF-Bipolar Cells of the Rat Retina. Cell Mol Neurobiol 2021; 41:229-245. [PMID: 32323153 PMCID: PMC7870642 DOI: 10.1007/s10571-020-00845-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/08/2020] [Indexed: 01/31/2023]
Abstract
Gap junctions are ubiquitous within the retina, but in general, it remains to be determined whether gap junction coupling between specific cell types is sufficiently strong to mediate functionally relevant coupling via electrical synapses. From ultrastructural, tracer coupling and immunolabeling studies, there is clear evidence for gap junctions between cone bipolar cells, but it is not known if these gap junctions function as electrical synapses. Here, using whole-cell voltage-clamp recording in rat (male and female) retinal slices, we investigated whether the gap junctions of bipolar cells make a measurable contribution to the membrane properties of these cells. We measured the input resistance (RN) of bipolar cells before and after applying meclofenamic acid (MFA) to block gap junctions. In the presence of MFA, RN of ON-cone bipolar cells displayed a clear increase, paralleled by block of the electrical coupling between these cells and AII amacrine cells in recordings of coupled cell pairs. For OFF-cone and rod bipolar cells, RN did not increase in the presence of MFA. The results for rod bipolar cells are consistent with the lack of gap junctions in these cells. However, for OFF-cone bipolar cells, our results suggest that the morphologically identified gap junctions between these cells do not support a junctional conductance that is sufficient to mediate effective electrical coupling. Instead, these junctions might play a role in chemical and/or metabolic coupling between subcellular compartments.
Collapse
Affiliation(s)
- Rémi Fournel
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway
| | - Espen Hartveit
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway.
| | - Margaret Lin Veruki
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway.
| |
Collapse
|
15
|
Olejnickova V, Kocka M, Kvasilova A, Kolesova H, Dziacky A, Gidor T, Gidor L, Sankova B, Gregorovicova M, Gourdie RG, Sedmera D. Gap Junctional Communication via Connexin43 between Purkinje Fibers and Working Myocytes Explains the Epicardial Activation Pattern in the Postnatal Mouse Left Ventricle. Int J Mol Sci 2021; 22:2475. [PMID: 33804428 PMCID: PMC7957598 DOI: 10.3390/ijms22052475] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/19/2021] [Accepted: 02/25/2021] [Indexed: 12/31/2022] Open
Abstract
The mammalian ventricular myocardium forms a functional syncytium due to flow of electrical current mediated in part by gap junctions localized within intercalated disks. The connexin (Cx) subunit of gap junctions have direct and indirect roles in conduction of electrical impulse from the cardiac pacemaker via the cardiac conduction system (CCS) to working myocytes. Cx43 is the dominant isoform in these channels. We have studied the distribution of Cx43 junctions between the CCS and working myocytes in a transgenic mouse model, which had the His-Purkinje portion of the CCS labeled with green fluorescence protein. The highest number of such connections was found in a region about one-third of ventricular length above the apex, and it correlated with the peak proportion of Purkinje fibers (PFs) to the ventricular myocardium. At this location, on the septal surface of the left ventricle, the insulated left bundle branch split into the uninsulated network of PFs that continued to the free wall anteriorly and posteriorly. The second peak of PF abundance was present in the ventricular apex. Epicardial activation maps correspondingly placed the site of the first activation in the apical region, while some hearts presented more highly located breakthrough sites. Taken together, these results increase our understanding of the physiological pattern of ventricular activation and its morphological underpinning through detailed CCS anatomy and distribution of its gap junctional coupling to the working myocardium.
Collapse
Affiliation(s)
- Veronika Olejnickova
- Institute of Anatomy, First Faculty of Medicine, Charles University, 128 00 Prague, Czech Republic; (V.O.); (M.K.); (A.K.); (H.K.); (A.D.); (T.G.); (L.G.); (B.S.); (M.G.)
- Institute of Physiology, CAS, 142 20 Prague, Czech Republic
| | - Matej Kocka
- Institute of Anatomy, First Faculty of Medicine, Charles University, 128 00 Prague, Czech Republic; (V.O.); (M.K.); (A.K.); (H.K.); (A.D.); (T.G.); (L.G.); (B.S.); (M.G.)
| | - Alena Kvasilova
- Institute of Anatomy, First Faculty of Medicine, Charles University, 128 00 Prague, Czech Republic; (V.O.); (M.K.); (A.K.); (H.K.); (A.D.); (T.G.); (L.G.); (B.S.); (M.G.)
| | - Hana Kolesova
- Institute of Anatomy, First Faculty of Medicine, Charles University, 128 00 Prague, Czech Republic; (V.O.); (M.K.); (A.K.); (H.K.); (A.D.); (T.G.); (L.G.); (B.S.); (M.G.)
- Institute of Physiology, CAS, 142 20 Prague, Czech Republic
| | - Adam Dziacky
- Institute of Anatomy, First Faculty of Medicine, Charles University, 128 00 Prague, Czech Republic; (V.O.); (M.K.); (A.K.); (H.K.); (A.D.); (T.G.); (L.G.); (B.S.); (M.G.)
- Department of Pediatric Cardiology, Motol University Hospital, 150 06 Prague, Czech Republic
| | - Tom Gidor
- Institute of Anatomy, First Faculty of Medicine, Charles University, 128 00 Prague, Czech Republic; (V.O.); (M.K.); (A.K.); (H.K.); (A.D.); (T.G.); (L.G.); (B.S.); (M.G.)
| | - Lihi Gidor
- Institute of Anatomy, First Faculty of Medicine, Charles University, 128 00 Prague, Czech Republic; (V.O.); (M.K.); (A.K.); (H.K.); (A.D.); (T.G.); (L.G.); (B.S.); (M.G.)
| | - Barbora Sankova
- Institute of Anatomy, First Faculty of Medicine, Charles University, 128 00 Prague, Czech Republic; (V.O.); (M.K.); (A.K.); (H.K.); (A.D.); (T.G.); (L.G.); (B.S.); (M.G.)
| | - Martina Gregorovicova
- Institute of Anatomy, First Faculty of Medicine, Charles University, 128 00 Prague, Czech Republic; (V.O.); (M.K.); (A.K.); (H.K.); (A.D.); (T.G.); (L.G.); (B.S.); (M.G.)
- Institute of Physiology, CAS, 142 20 Prague, Czech Republic
| | - Robert G. Gourdie
- Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA 24016, USA;
| | - David Sedmera
- Institute of Anatomy, First Faculty of Medicine, Charles University, 128 00 Prague, Czech Republic; (V.O.); (M.K.); (A.K.); (H.K.); (A.D.); (T.G.); (L.G.); (B.S.); (M.G.)
- Institute of Physiology, CAS, 142 20 Prague, Czech Republic
| |
Collapse
|
16
|
Zhu Q, Yang G, Chen B, Liu F, Li X, Liu L. Altered Expression of GJD2 Messenger RNA and the Coded Protein Connexin 36 in Negative Lens-induced Myopia of Guinea Pigs. Optom Vis Sci 2020; 97:1080-1088. [PMID: 33278187 PMCID: PMC7742206 DOI: 10.1097/opx.0000000000001611] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 07/30/2020] [Indexed: 02/05/2023] Open
Abstract
SIGNIFICANCE Decreased expression of the retinal GJD2 gene messenger RNA (mRNA) and connexin 36 (Cx36) protein in the guinea pig negative lens-induced myopia (LIM) model suggests their involvement in local retinal circuits regulating eye growth. PURPOSE Previous studies suggest that the GJD2 gene and Cx36 protein encoded by the GJD2 gene play important roles in retinal signaling pathways and eye development. The aim of this study was to investigate the changes in GJD2 mRNA and Cx36 protein expression in the guinea pig lens-induced myopia model. METHODS Four-week-old guinea pigs were randomly divided into two groups. Animals in the experimental group were fitted with monocular -10 D lenses; and animals in the control group, with monocular plano lenses. Biometric measurements, including the spherical equivalent refractive error and axial length, were monitored. Animals were killed after 0, 1, 2, and 3 weeks of treatment, and their retinas were isolated. Retinal GJD2 mRNA and Cx36 protein expression levels were assessed by quantitative real-time polymerase chain reaction and Western blot analysis, respectively. RESULTS Spherical equivalent refractive error values indicated that negative lens-treated eyes became significantly more myopic than plano lens-treated eyes (P = .001), consistent with their longer axial lengths compared with those of control eyes. Both GJD2 mRNA and Cx36 protein expression levels were decreased in the retinas of negative lens-treated eyes compared with levels in the retinas of plano lens-treated eyes, although there were differences in the timing; GJD2 mRNA, levels were significantly decreased after 1 and 2 weeks of treatment (P = .01 and P = .004, respectively), whereas Cx36 protein expression was significantly decreased after only 1 week (P = .01). CONCLUSIONS That both retinal GJD2 mRNA and Cx36 protein expression levels were decreased after induction of myopia with negative lenses points to retinal circuits involving Cx36 in myopia development in the guinea pig.
Collapse
Affiliation(s)
- Qiurong Zhu
- Department of Optometry and Visual Science, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Guoyuan Yang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bingjie Chen
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fengyang Liu
- Department of Optometry, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Xia Li
- Department of Optometry and Visual Science, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Longqian Liu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
17
|
Smedowski A, Akhtar S, Liu X, Pietrucha‐Dutczak M, Podracka L, Toropainen E, Alkanaan A, Ruponen M, Urtti A, Varjosalo M, Kaarniranta K, Lewin‐Kowalik J. Electrical synapses interconnecting axons revealed in the optic nerve head - a novel model of gap junctions' involvement in optic nerve function. Acta Ophthalmol 2020; 98:408-417. [PMID: 31602808 PMCID: PMC7318195 DOI: 10.1111/aos.14272] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 09/14/2019] [Indexed: 01/14/2023]
Abstract
PURPOSE To characterize newly discovered electrical synapses, formed by connexin (Cx) 36 and 45, between neighbouring axons within the optic nerve head. METHODS Twenty-five Wistar rats were killed by CO2 inhalation. Proximal and distal optic nerve (ON) stumps were collected and processed for immunostainings, electron microscopy (EM) with immunogold labelling, PCR and Western blots (WB). Additional 15 animals were deeply anaesthetized, and flash visual evoked potentials (fVEP) after retrobulbar injection of saline (negative control) or 100 μm meclofenamic acid solution (gap junctions' blocker) were recorded. Human paraffin cross-sections of eyeballs for immunostainings were obtained from the Human Eye Biobank for Research. RESULTS Immunostainings of both rat and human ON revealed the presence of Cx45 and 36 colocalizing with β3-tubulin, but not with glial fibrillary acidic protein (GFAP). In WB, Cx36 content in optic nerve was approximately halved when compared with retina (0.58 ± 0.005 in proximal stump and 0.44 ± 0.02 in distal stump), Cx45 showed higher levels (0.68 ± 0.01 in proximal stump and 0.9 ± 0.07 in distal stump). In immunogold-EM of optic nerve sections, we found electric synapses (formed mostly by Cx45) directly coupling neighbouring axons. In fVEP, blocking of gap junctions with meclofenamic acid resulted in significant prolongation of the latency of P1 wave up to 160% after 30 min (p < 0.001). CONCLUSIONS Optic nerve (ON) axons are equipped with electrical synapses composed of neuronal connexins, especially Cx45, creating direct morphological and functional connections between each other. This finding could have substantial implications for understanding of the pathogenesis of various optic neuropathies and identifies a new potential target for a therapeutic approach.
Collapse
Affiliation(s)
- Adrian Smedowski
- Department of PhysiologySchool of Medicine in KatowiceMedical University of SilesiaKatowicePoland
| | - Saeed Akhtar
- Department of OptometryCollege of Applied Medical SciencesKing Saud UniversityRiyadhKingdom of Saudi Arabia
| | - Xiaonan Liu
- Institute of BiotechnologyUniversity of HelsinkiHelsinkiFinland
| | - Marita Pietrucha‐Dutczak
- Department of PhysiologySchool of Medicine in KatowiceMedical University of SilesiaKatowicePoland
| | - Lucia Podracka
- School of PharmacyUniversity of Eastern FinlandKuopioFinland
| | | | - Aljoharah Alkanaan
- Department of OptometryCollege of Applied Medical SciencesKing Saud UniversityRiyadhKingdom of Saudi Arabia
| | - Marika Ruponen
- School of PharmacyUniversity of Eastern FinlandKuopioFinland
| | - Arto Urtti
- School of PharmacyUniversity of Eastern FinlandKuopioFinland
| | | | - Kai Kaarniranta
- Department of OphthalmologyUniversity of Eastern FinlandKuopioFinland,Department of OphthalmologyKuopio University HospitalKuopioFinland
| | - Joanna Lewin‐Kowalik
- Department of PhysiologySchool of Medicine in KatowiceMedical University of SilesiaKatowicePoland
| |
Collapse
|
18
|
Network Architecture of Gap Junctional Coupling among Parallel Processing Channels in the Mammalian Retina. J Neurosci 2020; 40:4483-4511. [PMID: 32332119 DOI: 10.1523/jneurosci.1810-19.2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 03/27/2020] [Accepted: 04/12/2020] [Indexed: 01/04/2023] Open
Abstract
Gap junctions are ubiquitous throughout the nervous system, mediating critical signal transmission and integration, as well as emergent network properties. In mammalian retina, gap junctions within the Aii amacrine cell-ON cone bipolar cell (CBC) network are essential for night vision, modulation of day vision, and contribute to visual impairment in retinal degenerations, yet neither the extended network topology nor its conservation is well established. Here, we map the network contribution of gap junctions using a high-resolution connectomics dataset of an adult female rabbit retina. Gap junctions are prominent synaptic components of ON CBC classes, constituting 5%-25% of all axonal synaptic contacts. Many of these mediate canonical transfer of rod signals from Aii cells to ON CBCs for night vision, and we find that the uneven distribution of Aii signals to ON CBCs is conserved in rabbit, including one class entirely lacking direct Aii coupling. However, the majority of gap junctions formed by ON CBCs unexpectedly occur between ON CBCs, rather than with Aii cells. Such coupling is extensive, creating an interconnected network with numerous lateral paths both within, and particularly across, these parallel processing streams. Coupling patterns are precise with ON CBCs accepting and rejecting unique combinations of partnerships according to robust rulesets. Coupling specificity extends to both size and spatial topologies, thereby rivaling the synaptic specificity of chemical synapses. These ON CBC coupling motifs dramatically extend the coupled Aii-ON CBC network, with implications for signal flow in both scotopic and photopic retinal networks during visual processing and disease.SIGNIFICANCE STATEMENT Electrical synapses mediated by gap junctions are fundamental components of neural networks. In retina, coupling within the Aii-ON CBC network shapes visual processing in both the scotopic and photopic networks. In retinal degenerations, these same gap junctions mediate oscillatory activity that contributes to visual impairment. Here, we use high-resolution connectomics strategies to identify gap junctions and cellular partnerships. We describe novel, pervasive motifs both within and across classes of ON CBCs that dramatically extend the Aii-ON CBC network. These motifs are highly specific with implications for both signal processing within the retina and therapeutic interventions for blinding conditions. These findings highlight the underappreciated contribution of coupling motifs in retinal circuitry and the necessity of their detection in connectomics studies.
Collapse
|
19
|
Appleby TR, Manookin MB. Selectivity to approaching motion in retinal inputs to the dorsal visual pathway. eLife 2020; 9:e51144. [PMID: 32091390 PMCID: PMC7080407 DOI: 10.7554/elife.51144] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 02/18/2020] [Indexed: 11/13/2022] Open
Abstract
To efficiently navigate through the environment and avoid potential threats, an animal must quickly detect the motion of approaching objects. Current models of primate vision place the origins of this complex computation in the visual cortex. Here, we report that detection of approaching motion begins in the retina. Several ganglion cell types, the retinal output neurons, show selectivity to approaching motion. Synaptic current recordings from these cells further reveal that this preference for approaching motion arises in the interplay between presynaptic excitatory and inhibitory circuit elements. These findings demonstrate how excitatory and inhibitory circuits interact to mediate an ethologically relevant neural function. Moreover, the elementary computations that detect approaching motion begin early in the visual stream of primates.
Collapse
Affiliation(s)
- Todd R Appleby
- Graduate Program in Neuroscience, University of WashingtonSeattleUnited States
- Department of Ophthalmology, University of WashingtonSeattleUnited States
- Vision Science Center, University of WashingtonSeattleUnited States
| | - Michael B Manookin
- Department of Ophthalmology, University of WashingtonSeattleUnited States
- Vision Science Center, University of WashingtonSeattleUnited States
| |
Collapse
|
20
|
Ivanova E, Kovacs-Oller T, Sagdullaev BT. Domain-specific distribution of gap junctions defines cellular coupling to establish a vascular relay in the retina. J Comp Neurol 2019; 527:2675-2693. [PMID: 30950036 PMCID: PMC6721971 DOI: 10.1002/cne.24699] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/02/2019] [Accepted: 04/02/2019] [Indexed: 12/22/2022]
Abstract
In the retina, diverse functions of neuronal gap junctions (GJs) have been established. However, the distribution and function of vascular GJs are less clear. Here in the mouse retina whole mounts, we combined structural immunohistochemical analysis and a functional assessment of cellular coupling with a GJ-permeable tracer Neurobiotin to determine distribution patterns of three major vascular connexins. We found that Cx43 was expressed in punctate fashion on astroglia, surrounding all types of blood vessels and in continuous string-like structures along endothelial cell contacts in specialized regions of the vascular tree. Specifically, these Cx43-positive strings originated at the finest capillaries and extended toward the feeding artery. As this structural arrangement promoted strong and exclusive coupling of pericytes and endothelial cells along the corresponding branch, we termed this region a "vascular relay." Cx40 expression was found predominantly along the endothelial cell contacts of the primary arteries and did not overlap with Cx43-positive strings. At their occupied territories, Cx43 and Cx40 clustered with tight junctions and, to a lesser extent, with adhesion contacts, both key elements of the blood-retina barrier. Finally, Cx37 puncta were associated with the entire surface of both mural and endothelial cells across all regions of the vascular tree. This combinatorial analysis of vascular connexins and identification of the vascular relay region will serve as a structural foundation for future studies of neurovascular signaling in health and disease.
Collapse
Affiliation(s)
- Elena Ivanova
- Burke Neurological Institute, Department of Ophthalmology, Weill Cornell Medicine, White Plains, New York
| | - Tamas Kovacs-Oller
- Burke Neurological Institute, Department of Ophthalmology, Weill Cornell Medicine, White Plains, New York
| | - Botir T Sagdullaev
- Burke Neurological Institute, Department of Ophthalmology, Weill Cornell Medicine, White Plains, New York
| |
Collapse
|
21
|
Matsumoto A, Tachibana M. Global Jitter Motion of the Retinal Image Dynamically Alters the Receptive Field Properties of Retinal Ganglion Cells. Front Neurosci 2019; 13:979. [PMID: 31572123 PMCID: PMC6753181 DOI: 10.3389/fnins.2019.00979] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 08/30/2019] [Indexed: 11/25/2022] Open
Abstract
Fixational eye movements induce aperiodic motion of the retinal image. However, it is not yet fully understood how fixational eye movements affect retinal information processing. Here we show that global jitter motion, simulating the image motion during fixation, alters the spatiotemporal receptive field properties of retinal ganglion cells. Using multi-electrode and whole-cell recording techniques, we investigated light-evoked responses from ganglion cells in the isolated goldfish retina. Ganglion cells were classified into six groups based on the filtering property of light stimulus, the membrane properties, and the cell morphology. The spatiotemporal receptive field profiles of retinal ganglion cells were estimated by the reverse correlation method, where the dense noise stimulus was applied on the dark or random-dot background. We found that the jitter motion of the random-dot background elongated the receptive filed along the rostral-caudal axis and temporally sensitized in a specific group of ganglion cells: Fast-transient ganglion cells. At the newly emerged regions of the receptive field local light stimulation evoked excitatory postsynaptic currents with large amplitude and fast kinetics without changing the properties of inhibitory postsynaptic currents. Pharmacological experiments suggested two presynaptic mechanisms underlying the receptive field alteration: (i) electrical coupling between bipolar cells, which expands the receptive field in all directions; (ii) GABAergic presynaptic inhibition from amacrine cells, which reduces the dorsal and ventral regions of the expanded receptive field, resulting in elongation along the rostral-caudal axis. Our study demonstrates that the receptive field of Fast-transient ganglion cells is not static but dynamically altered depending on the visual inputs. The receptive field elongation during fixational eye movements may contribute to prompt firing to a target in the succeeding saccade.
Collapse
Affiliation(s)
- Akihiro Matsumoto
- Department of Psychology, Graduate School of Humanities and Sociology, The University of Tokyo, Tokyo, Japan
- Ritsumeikan Global Innovation Research Organization (R-GIRO), Ritsumeikan University, Kusatsu, Japan
- Danish Research Institute of Translational Neuroscience (DANDRITE), Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Masao Tachibana
- Department of Psychology, Graduate School of Humanities and Sociology, The University of Tokyo, Tokyo, Japan
- Research Organization of Science and Technology, Ritsumeikan University, Kusatsu, Japan
| |
Collapse
|
22
|
Telkes I, Kóbor P, Orbán J, Kovács-Öller T, Völgyi B, Buzás P. Connexin-36 distribution and layer-specific topography in the cat retina. Brain Struct Funct 2019; 224:2183-2197. [PMID: 31172263 PMCID: PMC6591202 DOI: 10.1007/s00429-019-01876-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 04/11/2019] [Indexed: 11/29/2022]
Abstract
Connexin-36 (Cx36) is the major constituent of mammalian retinal gap junctions positioned in key signal pathways. Here, we examined the laminar and large-scale topographical distribution of Cx36 punctate immunolabels in the retina of the cat, a classical model of the mammalian visual system. Calretinin-immunoreactive (CaR-IR) cell populations served to outline the nuclear and plexiform layers and to stain specific neuronal populations. CaR-IR cells included horizontal cells in the outer retina, numerous amacrine cells, and scattered cells in the ganglion cell layer. Cx36-IR plaques were found among horizontal cell dendrites albeit without systematic colocalization of the two labels. Diffuse Cx36 immunoreactivity was found in the cytoplasm of AII amacrine cells, but no colocalization of Cx36 plaques was observed with either the perikarya or the long varicose dendrites of the CaR-IR non-AII amacrine cells. Cx36 puncta were seen throughout the entire inner plexiform layer showing their highest density in the ON sublamina. The densities of AII amacrine cell bodies and Cx36 plaques in the ON sublamina were strongly correlated across a wide range of eccentricities suggesting their anatomical association. However, the high number of plaques per AII cell suggests that a considerable fraction of Cx36 gap junctions in the ON sublamina is formed by other cell types than AII amacrine cells drawing attention to extensive but less studied electrically coupled networks.
Collapse
Affiliation(s)
- Ildikó Telkes
- Institute of Physiology, Medical School, University of Pécs, Szigeti út 12, Pécs, 7624, Hungary
- Szentágothai Research Centre, University of Pécs, Pécs, 7624, Hungary
- Centre for Neuroscience, University of Pécs, Pécs, 7624, Hungary
| | - Péter Kóbor
- Institute of Physiology, Medical School, University of Pécs, Szigeti út 12, Pécs, 7624, Hungary
- Szentágothai Research Centre, University of Pécs, Pécs, 7624, Hungary
- Centre for Neuroscience, University of Pécs, Pécs, 7624, Hungary
| | - József Orbán
- Szentágothai Research Centre, University of Pécs, Pécs, 7624, Hungary
- Department of Biophysics, Medical School, University of Pécs, Pécs, 7624, Hungary
| | - Tamás Kovács-Öller
- Szentágothai Research Centre, University of Pécs, Pécs, 7624, Hungary
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, 7624, Hungary
- Retinal Electrical Synapses Research Group, MTA-PTE NAP-2, University of Pécs, Pécs, 7624, Hungary
- Centre for Neuroscience, University of Pécs, Pécs, 7624, Hungary
| | - Béla Völgyi
- Szentágothai Research Centre, University of Pécs, Pécs, 7624, Hungary
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, 7624, Hungary
- Retinal Electrical Synapses Research Group, MTA-PTE NAP-2, University of Pécs, Pécs, 7624, Hungary
- Centre for Neuroscience, University of Pécs, Pécs, 7624, Hungary
| | - Péter Buzás
- Institute of Physiology, Medical School, University of Pécs, Szigeti út 12, Pécs, 7624, Hungary.
- Szentágothai Research Centre, University of Pécs, Pécs, 7624, Hungary.
- Centre for Neuroscience, University of Pécs, Pécs, 7624, Hungary.
| |
Collapse
|
23
|
Kovács-Öller T, Szarka G, Ganczer A, Tengölics Á, Balogh B, Völgyi B. Expression of Ca 2+-Binding Buffer Proteins in the Human and Mouse Retinal Neurons. Int J Mol Sci 2019; 20:E2229. [PMID: 31067641 PMCID: PMC6539911 DOI: 10.3390/ijms20092229] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 12/31/2022] Open
Abstract
Ca2+-binding buffer proteins (CaBPs) are widely expressed by various neurons throughout the central nervous system (CNS), including the retina. While the expression of CaBPs by photoreceptors, retinal interneurons and the output ganglion cells in the mammalian retina has been extensively studied, a general description is still missing due to the differences between species, developmental expression patterns and study-to-study discrepancies. Furthermore, CaBPs are occasionally located in a compartment-specific manner and two or more CaBPs can be expressed by the same neuron, thereby sharing the labor of Ca2+ buffering in the intracellular milieu. This article reviews this topic by providing a framework on CaBP functional expression by neurons of the mammalian retina with an emphasis on human and mouse retinas and the three most abundant and extensively studied buffer proteins: parvalbumin, calretinin and calbindin.
Collapse
Affiliation(s)
- Tamás Kovács-Öller
- János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary.
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, 1051 Budapest, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
- Medical School, University of Pécs, 7624 Pécs, Hungary.
| | - Gergely Szarka
- János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary.
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, 1051 Budapest, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
| | - Alma Ganczer
- János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary.
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, 1051 Budapest, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
| | - Ádám Tengölics
- János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary.
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, 1051 Budapest, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
| | - Boglárka Balogh
- János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary.
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, 1051 Budapest, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
| | - Béla Völgyi
- János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary.
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, 1051 Budapest, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
- Medical School, University of Pécs, 7624 Pécs, Hungary.
| |
Collapse
|
24
|
Roy S, Field GD. Dopaminergic modulation of retinal processing from starlight to sunlight. J Pharmacol Sci 2019; 140:86-93. [PMID: 31109761 DOI: 10.1016/j.jphs.2019.03.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/13/2019] [Accepted: 03/29/2019] [Indexed: 12/17/2022] Open
Abstract
Neuromodulators such as dopamine, enable context-dependent plasticity of neural circuit function throughout the central nervous system. For example, in the retina, dopamine tunes visual processing for daylight and nightlight conditions. Specifically, high levels of dopamine release in the retina tune vision for daylight (photopic) conditions, while low levels tune it for nightlight (scotopic) conditions. This review covers the cellular and circuit-level mechanisms within the retina that are altered by dopamine. These mechanisms include changes in gap junction coupling and ionic conductances, both of which are altered by the activation of diverse types of dopamine receptors across diverse types of retinal neurons. We contextualize the modulatory actions of dopamine in terms of alterations and optimizations to visual processing under photopic and scotopic conditions, with particular attention to how they differentially impact distinct cell types. Finally, we discuss how transgenic mice and disease models have shaped our understanding of dopaminergic signaling and its role in visual processing. Cumulatively, this review illustrates some of the diverse and potent mechanisms through which neuromodulation can shape brain function.
Collapse
Affiliation(s)
- Suva Roy
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, USA
| | - Greg D Field
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
25
|
Kántor O, Szarka G, Benkő Z, Somogyvári Z, Pálfi E, Baksa G, Rácz G, Nitschke R, Debertin G, Völgyi B. Strategic Positioning of Connexin36 Gap Junctions Across Human Retinal Ganglion Cell Dendritic Arbors. Front Cell Neurosci 2018; 12:409. [PMID: 30524239 PMCID: PMC6262005 DOI: 10.3389/fncel.2018.00409] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/22/2018] [Indexed: 11/18/2022] Open
Abstract
Connexin36 (Cx36) subunits form gap junctions (GJ) between neurons throughout the central nervous system. Such GJs of the mammalian retina serve the transmission, averaging and correlation of signals prior to conveying visual information to the brain. Retinal GJs have been exhaustively studied in various animal species, however, there is still a perplexing paucity of information regarding the presence and function of human retinal GJs. Particularly little is known about GJ formation of human retinal ganglion cells (hRGCs) due to the limited number of suitable experimental approaches. Compared to the neuronal coupling studies in animal models, where GJ permeable tracer injection is the gold standard method, the post-mortem nature of scarcely available human retinal samples leaves immunohistochemistry as a sole approach to obtain information on hRGC GJs. In this study Lucifer Yellow (LY) dye injections and Cx36 immunohistochemistry were performed in fixed short-post-mortem samples to stain hRGCs with complete dendritic arbors and locate dendritic Cx36 GJs. Subsequent neuronal reconstructions and morphometric analyses revealed that Cx36 plaques had a clear tendency to form clusters and particularly favored terminal dendritic segments.
Collapse
Affiliation(s)
- Orsolya Kántor
- Department of Neuroanatomy, Faculty of Medicine, Institute for Anatomy and Cell Biology, Albert-Ludwigs-University Freiburg, Freiburg, Germany.,MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, Pécs, Hungary.,Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Gergely Szarka
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, Pécs, Hungary.,Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary.,Center for Neuroscience, University of Pécs, Pécs, Hungary.,János Szentágothai Research Center, University of Pécs, Pécs, Hungary
| | - Zsigmond Benkő
- Complex Systems and Computational Neuroscience Group, Wigner Research Center for Physics, Hungarian Academy of Sciences, Budapest, Hungary
| | - Zoltán Somogyvári
- Complex Systems and Computational Neuroscience Group, Wigner Research Center for Physics, Hungarian Academy of Sciences, Budapest, Hungary
| | - Emese Pálfi
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Gábor Baksa
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Gergely Rácz
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Roland Nitschke
- Life Imaging Center, Center for Biological Systems Analysis, Albert-Ludwigs University, Freiburg, Germany.,BIOSS Center for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Gábor Debertin
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, Pécs, Hungary.,Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary.,Center for Neuroscience, University of Pécs, Pécs, Hungary.,János Szentágothai Research Center, University of Pécs, Pécs, Hungary
| | - Béla Völgyi
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, Pécs, Hungary.,Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary.,Center for Neuroscience, University of Pécs, Pécs, Hungary.,János Szentágothai Research Center, University of Pécs, Pécs, Hungary
| |
Collapse
|
26
|
Marc RE, Sigulinsky CL, Pfeiffer RL, Emrich D, Anderson JR, Jones BW. Heterocellular Coupling Between Amacrine Cells and Ganglion Cells. Front Neural Circuits 2018; 12:90. [PMID: 30487737 PMCID: PMC6247779 DOI: 10.3389/fncir.2018.00090] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 09/28/2018] [Indexed: 01/08/2023] Open
Abstract
All superclasses of retinal neurons, including bipolar cells (BCs), amacrine cells (ACs) and ganglion cells (GCs), display gap junctional coupling. However, coupling varies extensively by class. Heterocellular AC coupling is common in many mammalian GC classes. Yet, the topology and functions of coupling networks remains largely undefined. GCs are the least frequent superclass in the inner plexiform layer and the gap junctions mediating GC-to-AC coupling (GC::AC) are sparsely arrayed amidst large cohorts of homocellular AC::AC, BC::BC, GC::GC and heterocellular AC::BC gap junctions. Here, we report quantitative coupling for identified GCs in retinal connectome 1 (RC1), a high resolution (2 nm) transmission electron microscopy-based volume of rabbit retina. These reveal that most GC gap junctions in RC1 are suboptical. GC classes lack direct cross-class homocellular coupling with other GCs, despite opportunities via direct membrane contact, while OFF alpha GCs and transient ON directionally selective (DS) GCs are strongly coupled to distinct AC cohorts. Integrated small molecule immunocytochemistry identifies these as GABAergic ACs (γ+ ACs). Multi-hop synaptic queries of RC1 connectome further profile these coupled γ+ ACs. Notably, OFF alpha GCs couple to OFF γ+ ACs and transient ON DS GCs couple to ON γ+ ACs, including a large interstitial amacrine cell, revealing matched ON/OFF photic drive polarities within coupled networks. Furthermore, BC input to these γ+ ACs is tightly matched to the GCs with which they couple. Evaluation of the coupled versus inhibitory targets of the γ+ ACs reveals that in both ON and OFF coupled GC networks these ACs are presynaptic to GC classes that are different than the classes with which they couple. These heterocellular coupling patterns provide a potential mechanism for an excited GC to indirectly inhibit nearby GCs of different classes. Similarly, coupled γ+ ACs engaged in feedback networks can leverage the additional gain of BC synapses in shaping the signaling of downstream targets based on their own selective coupling with GCs. A consequence of coupling is intercellular fluxes of small molecules. GC::AC coupling involves primarily γ+ cells, likely resulting in GABA diffusion into GCs. Surveying GABA signatures in the GC layer across diverse species suggests the majority of vertebrate retinas engage in GC::γ+ AC coupling.
Collapse
Affiliation(s)
| | | | | | | | | | - Bryan William Jones
- Moran Eye Center, Department of Ophthalmology and Visual Sciences, The University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
27
|
O'Brien J, Bloomfield SA. Plasticity of Retinal Gap Junctions: Roles in Synaptic Physiology and Disease. Annu Rev Vis Sci 2018; 4:79-100. [DOI: 10.1146/annurev-vision-091517-034133] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Electrical synaptic transmission via gap junctions underlies direct and rapid neuronal communication in the central nervous system. The diversity of functional roles played by electrical synapses is perhaps best exemplified in the vertebrate retina, in which gap junctions are expressed by each of the five major neuronal types. These junctions are highly plastic; they are dynamically regulated by ambient illumination and circadian rhythms acting through light-activated neuromodulators. The networks formed by electrically coupled neurons provide plastic, reconfigurable circuits positioned to play key and diverse roles in the transmission and processing of visual information at every retinal level. Recent work indicates gap junctions also play a role in the progressive cell death and aberrant activity seen in various pathological conditions of the retina. Gap junctions thus form potential targets for novel neuroprotective therapies in the treatment of neurodegenerative retinal diseases such as glaucoma and ischemic retinopathies.
Collapse
Affiliation(s)
- John O'Brien
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center, Houston, Texas 77030, USA
| | - Stewart A. Bloomfield
- Department of Biological and Vision Sciences, State University of New York College of Optometry, New York, NY 10036, USA
| |
Collapse
|
28
|
An Alternative Splice Variant of Zebrafish Cx52.6 is Expressed in Retinal Horizontal Cells. Neuroscience 2018; 388:191-202. [PMID: 30048782 DOI: 10.1016/j.neuroscience.2018.07.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/27/2018] [Accepted: 07/16/2018] [Indexed: 11/22/2022]
Abstract
Retinal horizontal cells (HCs) are inhibitory neurons, which modulate the transmission of light-elicited signals from photoreceptors to bipolar cells in the outer retina. HCs of the same physiological type are extensively coupled via gap junctions. In the zebrafish retina, the population of HCs comprises up to four morphologically distinct subtypes. Four different connexins (Cx52.6, Cx52.7, Cx52.9 and Cx55.5) were detected in these cells with overlapping expression patterns. In this study, we show that Cx52.6 is alternatively spliced in the retina, resulting in an additional isoform, designated as Cx53.4, which differs from the originally described Cx52.6 only by the final C-terminal peptide (12 vs. 4 aa). Further protein sequence alignments revealed that Cx53.4 represents the counterpart of alternatively spliced mouse Cx57 and human Cx62. RT-PCR analyses of mRNA expression in different adult zebrafish tissues showed that Cx53.4 is expressed exclusively in the retina. The localization of Cx53.4 protein within the retina was analyzed using a specific antibody. Immunofluorescence analyses demonstrated that the expression of Cx53.4 is restricted to HCs of all four subtypes. Further, immunoelectron microscopy confirmed the presence of Cx53.4 in gap junctions between HC dendrites and between their axon terminals.
Collapse
|
29
|
Manookin MB, Patterson SS, Linehan CM. Neural Mechanisms Mediating Motion Sensitivity in Parasol Ganglion Cells of the Primate Retina. Neuron 2018; 97:1327-1340.e4. [PMID: 29503188 PMCID: PMC5866240 DOI: 10.1016/j.neuron.2018.02.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 01/16/2018] [Accepted: 02/02/2018] [Indexed: 10/17/2022]
Abstract
Considerable theoretical and experimental effort has been dedicated to understanding how neural circuits detect visual motion. In primates, much is known about the cortical circuits that contribute to motion processing, but the role of the retina in this fundamental neural computation is poorly understood. Here, we used a combination of extracellular and whole-cell recording to test for motion sensitivity in the two main classes of output neurons in the primate retina-midget (parvocellular-projecting) and parasol (magnocellular-projecting) ganglion cells. We report that parasol, but not midget, ganglion cells are motion sensitive. This motion sensitivity is present in synaptic excitation and disinhibition from presynaptic bipolar cells and amacrine cells, respectively. Moreover, electrical coupling between neighboring bipolar cells and the nonlinear nature of synaptic release contribute to the observed motion sensitivity. Our findings indicate that motion computations arise far earlier in the primate visual stream than previously thought.
Collapse
Affiliation(s)
- Michael B Manookin
- Department of Ophthalmology, University of Washington, Seattle, WA 98195, USA; Vision Science Center, University of Washington, Seattle, WA 98195, USA.
| | - Sara S Patterson
- Department of Ophthalmology, University of Washington, Seattle, WA 98195, USA; Vision Science Center, University of Washington, Seattle, WA 98195, USA; Graduate Program in Neuroscience, University of Washington, Seattle, WA 98195, USA
| | - Conor M Linehan
- Department of Ophthalmology, University of Washington, Seattle, WA 98195, USA; Vision Science Center, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
30
|
Elgueta C, Leroy F, Vielma AH, Schmachtenberg O, Palacios AG. Electrical coupling between A17 cells enhances reciprocal inhibitory feedback to rod bipolar cells. Sci Rep 2018; 8:3123. [PMID: 29449585 PMCID: PMC5814567 DOI: 10.1038/s41598-018-21119-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 01/29/2018] [Indexed: 11/09/2022] Open
Abstract
A17 amacrine cells are an important part of the scotopic pathway. Their synaptic varicosities receive glutamatergic inputs from rod bipolar cells (RBC) and release GABA onto the same RBC terminal, forming a reciprocal feedback that shapes RBC depolarization. Here, using patch-clamp recordings, we characterized electrical coupling between A17 cells of the rat retina and report the presence of strongly interconnected and non-coupled A17 cells. In coupled A17 cells, evoked currents preferentially flow out of the cell through GJs and cross-synchronization of presynaptic signals in a pair of A17 cells is correlated to their coupling degree. Moreover, we demonstrate that stimulation of one A17 cell can induce electrical and calcium transients in neighboring A17 cells, thus confirming a functional flow of information through electrical synapses in the A17 coupled network. Finally, blocking GJs caused a strong decrease in the amplitude of the inhibitory feedback onto RBCs. We therefore propose that electrical coupling between A17 cells enhances feedback onto RBCs by synchronizing and facilitating GABA release from inhibitory varicosities surrounding each RBC axon terminal. GJs between A17 cells are therefore critical in shaping the visual flow through the scotopic pathway.
Collapse
Affiliation(s)
- Claudio Elgueta
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile.
- Physiology Institute I, Alberts Ludwig University, Freiburg, Germany.
| | - Felix Leroy
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Neuroscience department, Columbia University Medical Center, 1051 Riverside Drive, New York, NY, 10032, USA
| | - Alex H Vielma
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Physiology Institute I, Alberts Ludwig University, Freiburg, Germany
| | - Oliver Schmachtenberg
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Physiology Institute I, Alberts Ludwig University, Freiburg, Germany
| | - Adrian G Palacios
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Physiology Institute I, Alberts Ludwig University, Freiburg, Germany
| |
Collapse
|
31
|
Tetenborg S, Yadav SC, Hormuzdi SG, Monyer H, Janssen-Bienhold U, Dedek K. Differential Distribution of Retinal Ca 2+/Calmodulin-Dependent Kinase II (CaMKII) Isoforms Indicates CaMKII-β and -δ as Specific Elements of Electrical Synapses Made of Connexin36 (Cx36). Front Mol Neurosci 2017; 10:425. [PMID: 29311815 PMCID: PMC5742114 DOI: 10.3389/fnmol.2017.00425] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 12/05/2017] [Indexed: 12/17/2022] Open
Abstract
AII amacrine cells are essential interneurons of the primary rod pathway and transmit rod-driven signals to ON cone bipolar cells to enable scotopic vision. Gap junctions made of connexin36 (Cx36) mediate electrical coupling among AII cells and between AII cells and ON cone bipolar cells. These gap junctions underlie a remarkable degree of plasticity and are modulated by different signaling cascades. In particular, Ca2+/calmodulin-dependent protein kinase II (CaMKII) has been characterized as an important regulator of Cx36, capable of potentiating electrical coupling in AII cells. However, it is unclear which CaMKII isoform mediates this effect. To obtain a more detailed understanding of the isoform composition of CaMKII at retinal gap junctions, we analyzed the retinal distribution of all four CaMKII isoforms using confocal microscopy. These experiments revealed a differential distribution of CaMKII isoforms: CaMKII-α was strongly expressed in starburst amacrine cells, which are known to lack electrical coupling. CaMKII-β was abundant in OFF bipolar cells, which form electrical synapses in the outer and the inner retina. CaMKII-γ was diffusely distributed across the entire retina and could not be assigned to a specific cell type. CaMKII-δ labeling was evident in bipolar and AII amacrine cells, which contain the majority of Cx36-immunoreactive puncta in the inner retina. We double-labeled retinas for Cx36 and the four CaMKII isoforms and revealed that the composition of the CaMKII enzyme differs between gap junctions in the outer and the inner retina: in the outer retina, only CaMKII-β colocalized with Cx36-containing gap junctions, whereas in the inner retina, CaMKII-β and -δ colocalized with Cx36. This finding suggests that gap junctions in the inner and the outer retina may be regulated differently although they both contain the same connexin. Taken together, our study identifies CaMKII-β and -δ as Cx36-specific regulators in the mouse retina with CaMKII-δ regulating the primary rod pathway.
Collapse
Affiliation(s)
- Stephan Tetenborg
- Animal Navigation/Neurosensorics, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany
| | - Shubhash C Yadav
- Animal Navigation/Neurosensorics, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany
| | - Sheriar G Hormuzdi
- Division of Neuroscience, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | | | - Ulrike Janssen-Bienhold
- Visual Neuroscience, Department of Neuroscience, University of Oldenburg, Oldenburg, Germany.,Research Center Neurosensory Science, University of Oldenburg, Oldenburg, Germany
| | - Karin Dedek
- Animal Navigation/Neurosensorics, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany.,Research Center Neurosensory Science, University of Oldenburg, Oldenburg, Germany
| |
Collapse
|
32
|
Kántor O, Varga A, Nitschke R, Naumann A, Énzsöly A, Lukáts Á, Szabó A, Németh J, Völgyi B. Bipolar cell gap junctions serve major signaling pathways in the human retina. Brain Struct Funct 2017; 222:2603-2624. [PMID: 28070649 DOI: 10.1007/s00429-016-1360-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 12/22/2016] [Indexed: 11/26/2022]
Abstract
Connexin36 (Cx36) constituent gap junctions (GJ) throughout the brain connect neurons into functional syncytia. In the retina they underlie the transmission, averaging and correlation of signals prior conveying visual information to the brain. This is the first study that describes retinal bipolar cell (BC) GJs in the human inner retina, whose function is enigmatic even in the examined animal models. Furthermore, a number of unique features (e.g. fovea, trichromacy, midget system) necessitate a reexamination of the animal model results in the human retina. Well-preserved postmortem human samples of this study are allowed to identify Cx36 expressing BCs neurochemically. Results reveal that both rod and cone pathway interneurons display strong Cx36 expression. Rod BC inputs to AII amacrine cells (AC) appear in juxtaposition to AII GJs, thus suggesting a strategic AII cell targeting by rod BCs. Cone BCs serving midget, parasol or koniocellular signaling pathways display a wealth of Cx36 expression to form homologously coupled arrays. In addition, they also establish heterologous GJ contacts to serve an exchange of information between parallel signaling streams. Interestingly, a prominent Cx36 expression was exhibited by midget system BCs that appear to maintain intimate contacts with bistratified BCs serving other pathways. These findings suggest that BC GJs in parallel signaling streams serve both an intra- and inter-pathway exchange of signals in the human retina.
Collapse
Affiliation(s)
- Orsolya Kántor
- Department of Neuroanatomy, Faculty of Medicine, Institute for Anatomy and Cell Biology, University of Freiburg, 79104, Freiburg, Germany
- MTA-PTE NAP B Retinal Electrical Synapses Research Group, Pécs, 7624, Hungary
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Alexandra Varga
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Roland Nitschke
- Life Imaging Center, Center for Biological Systems Analysis, University of Freiburg, 79104, Freiburg, Germany
- BIOSS Centre for Biological Signaling Studies, University of Freiburg, 79104, Freiburg, Germany
| | - Angela Naumann
- Life Imaging Center, Center for Biological Systems Analysis, University of Freiburg, 79104, Freiburg, Germany
- BIOSS Centre for Biological Signaling Studies, University of Freiburg, 79104, Freiburg, Germany
| | - Anna Énzsöly
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
- Department of Ophthalmology, Semmelweis University, Budapest, 1085, Hungary
| | - Ákos Lukáts
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Arnold Szabó
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - János Németh
- Department of Ophthalmology, Semmelweis University, Budapest, 1085, Hungary
| | - Béla Völgyi
- MTA-PTE NAP B Retinal Electrical Synapses Research Group, Pécs, 7624, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, 7624, Hungary.
- János Szentágothai Research Center, University of Pécs, Ifjúság street 20, Pécs, 7624, Hungary.
- Department of Ophthalmology, New York University Langone Medical Center, New York, NY, 10016, USA.
| |
Collapse
|
33
|
Hou B, Fu Y, Weng C, Liu W, Zhao C, Yin ZQ. Homeostatic Plasticity Mediated by Rod-Cone Gap Junction Coupling in Retinal Degenerative Dystrophic RCS Rats. Front Cell Neurosci 2017; 11:98. [PMID: 28473754 PMCID: PMC5397418 DOI: 10.3389/fncel.2017.00098] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 03/22/2017] [Indexed: 11/14/2022] Open
Abstract
Rod-cone gap junctions open at night to allow rod signals to pass to cones and activate the cone-bipolar pathway. This enhances the ability to detect large, dim objects at night. This electrical synaptic switch is governed by the circadian clock and represents a novel form of homeostatic plasticity that regulates retinal excitability according to network activity. We used tracer labeling and ERG recording in the retinae of control and retinal degenerative dystrophic RCS rats. We found that in the control animals, rod-cone gap junction coupling was regulated by the circadian clock via the modulation of the phosphorylation of the melatonin synthetic enzyme arylalkylamine N-acetyltransferase (AANAT). However, in dystrophic RCS rats, AANAT was constitutively phosphorylated, causing rod-cone gap junctions to remain open. A further b/a-wave ratio analysis revealed that dystrophic RCS rats had stronger synaptic strength between photoreceptors and bipolar cells, possibly because rod-cone gap junctions remained open. This was despite the fact that a decrease was observed in the amplitude of both a- and b-waves as a result of the progressive loss of rods during early degenerative stages. These results suggest that electric synaptic strength is increased during the day to allow cone signals to pass to the remaining rods and to be propagated to rod bipolar cells, thereby partially compensating for the weak visual input caused by the loss of rods.
Collapse
Affiliation(s)
- Baoke Hou
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical UniversityChongqing, China.,Department of Ophthalmology, Chinese PLA General HospitalBeijing, China
| | - Yan Fu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical UniversityChongqing, China.,Key Lab of Visual Damage and Regeneration and Restoration of ChongqingChongqing, China
| | - Chuanhuang Weng
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical UniversityChongqing, China.,Key Lab of Visual Damage and Regeneration and Restoration of ChongqingChongqing, China
| | - Weiping Liu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical UniversityChongqing, China.,Key Lab of Visual Damage and Regeneration and Restoration of ChongqingChongqing, China
| | - Congjian Zhao
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical UniversityChongqing, China.,Key Lab of Visual Damage and Regeneration and Restoration of ChongqingChongqing, China
| | - Zheng Qin Yin
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical UniversityChongqing, China.,Key Lab of Visual Damage and Regeneration and Restoration of ChongqingChongqing, China
| |
Collapse
|
34
|
Connors BW. Synchrony and so much more: Diverse roles for electrical synapses in neural circuits. Dev Neurobiol 2017; 77:610-624. [PMID: 28245529 DOI: 10.1002/dneu.22493] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 02/05/2017] [Accepted: 02/14/2017] [Indexed: 11/09/2022]
Abstract
Electrical synapses are neuronal gap junctions that are ubiquitous across brain regions and species. The biophysical properties of most electrical synapses are relatively simple-transcellular channels allow nearly ohmic, bidirectional flow of ionic current. Yet these connections can play remarkably diverse roles in different neural circuit contexts. Recent findings illustrate how electrical synapses may excite or inhibit, synchronize or desynchronize, augment or diminish rhythms, phase-shift, detect coincidences, enhance signals relative to noise, adapt, and interact with nonlinear membrane and transmitter-release mechanisms. Most of these functions are likely to be widespread in central nervous systems. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 610-624, 2017.
Collapse
Affiliation(s)
- Barry W Connors
- Department of Neuroscience, Brown University, Providence, Rhode Island
| |
Collapse
|
35
|
Kovács-Öller T, Debertin G, Balogh M, Ganczer A, Orbán J, Nyitrai M, Balogh L, Kántor O, Völgyi B. Connexin36 Expression in the Mammalian Retina: A Multiple-Species Comparison. Front Cell Neurosci 2017; 11:65. [PMID: 28337128 PMCID: PMC5343066 DOI: 10.3389/fncel.2017.00065] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 02/23/2017] [Indexed: 11/23/2022] Open
Abstract
Much knowledge about interconnection of human retinal neurons is inferred from results on animal models. Likewise, there is a lack of information on human retinal electrical synapses/gap junctions (GJ). Connexin36 (Cx36) forms GJs in both the inner and outer plexiform layers (IPL and OPL) in most species including humans. However, a comparison of Cx36 GJ distribution in retinas of humans and popular animal models has not been presented. To this end a multiple-species comparison was performed in retinas of 12 mammals including humans to survey the Cx36 distribution. Areas of retinal specializations were avoided (e.g., fovea, visual streak, area centralis), thus observed Cx36 distribution differences were not attributed to these species-specific architecture of central retinal areas. Cx36 was expressed in both synaptic layers in all examined retinas. Cx36 plaques displayed an inhomogenous IPL distribution favoring the ON sublamina, however, this feature was more pronounced in the human, swine and guinea pig while it was less obvious in the rabbit, squirrel monkey, and ferret retinas. In contrast to the relative conservative Cx36 distribution in the IPL, the labels in the OPL varied considerably among mammals. In general, OPL plaques were rare and rather small in rod dominant carnivores and rodents, whereas the human and the cone rich guinea pig retinas displayed robust Cx36 labels. This survey presented that the human retina displayed two characteristic features, a pronounced ON dominance of Cx36 plaques in the IPL and prevalent Cx36 plaque conglomerates in the OPL. While many species showed either of these features, only the guinea pig retina shared both. The observed similarities and subtle differences in Cx36 plaque distribution across mammals do not correspond to evolutionary distances but may reflect accomodation to lifestyles of examined species.
Collapse
Affiliation(s)
- Tamás Kovács-Öller
- Department of Experimental Zoology and Neurobiology, University of PécsPécs, Hungary; János Szentágothai Research CenterPécs, Hungary; Retinal Electrical Synapses Research Group, Hungarian Academy of Sciences (MTA-PTE NAP B)Pécs, Hungary
| | - Gábor Debertin
- Department of Experimental Zoology and Neurobiology, University of PécsPécs, Hungary; János Szentágothai Research CenterPécs, Hungary; Retinal Electrical Synapses Research Group, Hungarian Academy of Sciences (MTA-PTE NAP B)Pécs, Hungary
| | - Márton Balogh
- Department of Experimental Zoology and Neurobiology, University of PécsPécs, Hungary; János Szentágothai Research CenterPécs, Hungary; Retinal Electrical Synapses Research Group, Hungarian Academy of Sciences (MTA-PTE NAP B)Pécs, Hungary
| | - Alma Ganczer
- Department of Experimental Zoology and Neurobiology, University of PécsPécs, Hungary; János Szentágothai Research CenterPécs, Hungary; Retinal Electrical Synapses Research Group, Hungarian Academy of Sciences (MTA-PTE NAP B)Pécs, Hungary
| | - József Orbán
- János Szentágothai Research CenterPécs, Hungary; Department of Biophysics, University of PécsPécs, Hungary; High-Field Terahertz Research Group, Hungarian Academy of Sciences (MTA-PTE)Pécs, Hungary
| | - Miklós Nyitrai
- János Szentágothai Research CenterPécs, Hungary; Department of Biophysics, University of PécsPécs, Hungary; Nuclear-Mitochondrial Interactions Research Group, Hungarian Academy of Sciences (MTA-PTE)Pécs, Hungary
| | - Lajos Balogh
- National Research Institute for Radiobiology and Radiohygiene Budapest, Hungary
| | - Orsolya Kántor
- Retinal Electrical Synapses Research Group, Hungarian Academy of Sciences (MTA-PTE NAP B)Pécs, Hungary; Department of Anatomy, Histology and Embryology, Semmelweis UniversityBudapest, Hungary; Department of Neuroanatomy, Institute for Anatomy and Cell Biology, Faculty of Medicine, University of FreiburgFreiburg, Germany
| | - Béla Völgyi
- Department of Experimental Zoology and Neurobiology, University of PécsPécs, Hungary; János Szentágothai Research CenterPécs, Hungary; Retinal Electrical Synapses Research Group, Hungarian Academy of Sciences (MTA-PTE NAP B)Pécs, Hungary; Department of Ophthalmology, New York University Langone Medical Center, New YorkNY, USA
| |
Collapse
|
36
|
Baker MW, Macagno ER. Gap junction proteins and the wiring (Rewiring) of neuronal circuits. Dev Neurobiol 2017; 77:575-586. [PMID: 27512961 DOI: 10.1002/dneu.22429] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 08/01/2016] [Accepted: 08/08/2016] [Indexed: 11/11/2022]
Abstract
The unique morphology and pattern of synaptic connections made by a neuron during development arise in part by an extended period of growth in which cell-cell interactions help to sculpt the arbor into its final shape, size, and participation in different synaptic networks. Recent experiments highlight a guiding role played by gap junction proteins in controlling this process. Ectopic and overexpression studies in invertebrates have revealed that the selective expression of distinct gap junction genes in neurons and glial cells is sufficient to establish selective new connections in the central nervous systems of the leech (Firme et al. [2012]: J Neurosci 32:14265-14270), the nematode (Rabinowitch et al. [2014]: Nat Commun 5:4442), and the fruit fly (Pézier et al., 2016: PLoS One 11:e0152211). We present here an overview of this work and suggest that gap junction proteins, in addition to their synaptic/communicative functions, have an instructive role as recognition and adhesion factors. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 575-586, 2017.
Collapse
Affiliation(s)
- Michael W Baker
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California, 92093
| | - Eduardo R Macagno
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California, 92093
| |
Collapse
|
37
|
MATSUMOTO A, TACHIBANA M. Rapid and coordinated processing of global motion images by local clusters of retinal ganglion cells. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2017; 93:234-249. [PMID: 28413199 PMCID: PMC5489431 DOI: 10.2183/pjab.93.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 02/14/2016] [Indexed: 06/07/2023]
Abstract
Even when the body is stationary, the whole retinal image is always in motion by fixational eye movements and saccades that move the eye between fixation points. Accumulating evidence indicates that the brain is equipped with specific mechanisms for compensating for the global motion induced by these eye movements. However, it is not yet fully understood how the retina processes global motion images during eye movements. Here we show that global motion images evoke novel coordinated firing in retinal ganglion cells (GCs). We simultaneously recorded the firing of GCs in the goldfish isolated retina using a multi-electrode array, and classified each GC based on the temporal profile of its receptive field (RF). A moving target that accompanied the global motion (simulating a saccade following a period of fixational eye movements) modulated the RF properties and evoked synchronized and correlated firing among local clusters of the specific GCs. Our findings provide a novel concept for retinal information processing during eye movements.
Collapse
Affiliation(s)
- Akihiro MATSUMOTO
- Department of Psychology, Graduate School of Humanities and Sociology, The University of Tokyo, Tokyo, Japan
| | - Masao TACHIBANA
- Department of Psychology, Graduate School of Humanities and Sociology, The University of Tokyo, Tokyo, Japan
- Center for Systems Vision Science, Organization of Science and Technology, Ritsumeikan University, Kusatsu, Shiga, Japan
| |
Collapse
|
38
|
Hidaka S. Conflicting effects by antibodies against connexin36 during the action of intracellular Cyclic-AMP onto electrical synapses of retinal ganglion cells. J Integr Neurosci 2016; 15:571-591. [PMID: 28052704 DOI: 10.1142/s021963521650031x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023] Open
Abstract
Alpha-type retinal ganglion cells (alpha cells) of the same class in mammalian retina are connected by gap junctions. Electrical synapses between alpha cells were examined using combined techniques of dual patch-clamp recordings, intracellular labeling and electron microscopy in the albino rat retina. In simultaneous dual whole-cell recordings from pairs of neighboring alpha cells, bidirectional electrical synapses with symmetrical junction conductance were observed in pairs with cells of the same morphological type. Regulatory domains of gap junction protein subunit connexins in electrical synapses between alpha cells by extracellular and intracellular ligands investigated by dual whole-patch clamp recordings. I examined how passage currents through electrical synapses between alpha cells are modulated by specific antibodies against connexin36 proteins, and extracellular or intracellular application of ligands. Control conditions led us to observe large passage currents between connected cells and adequate transjunctional conductance (Gj) (1.35[Formula: see text][Formula: see text][Formula: see text]0.51[Formula: see text]nS). Experimental results show that high level of intracellular cyclic AMP within examined cells suppress electrical synapses between the neighboring cells. Gj between examined cells reduced to 0.15[Formula: see text][Formula: see text][Formula: see text]0.04[Formula: see text]nS. Under application of dopamine (1.25[Formula: see text][Formula: see text][Formula: see text]0.06[Formula: see text]nS) or intracellular cyclic GMP (0.98[Formula: see text][Formula: see text][Formula: see text]0.23[Formula: see text]nS), however, Gj also remains as in the control level. Intracellular application of an antibody against the cytoplasmic loop of connexin36 reduced Gj (0.98[Formula: see text][Formula: see text][Formula: see text]0.23[Formula: see text]nS). Cocktail of the antibody against cytoplasmic connexin36 and intracellular cyclic AMP leaves Gj as in the level by single involvement of the cytoplasmic antibody. The elimination of Gj by the cytoplasmic antibody was in a dose-dependent manner. These results suggest that binding domains against cyclic AMP may be present in the cytoplasmic sites of connexin proteins to regulate channel opening of gap junctions between mammalian retinal alpha ganglion cells.
Collapse
Affiliation(s)
- Soh Hidaka
- 1 Department of Physiology, Fujita Health University School of Medicine, Toyoake Aichi 470-1192, Japan
| |
Collapse
|
39
|
Greb H, Hermann S, Dirks P, Ommen G, Kretschmer V, Schultz K, Zoidl G, Weiler R, Janssen-Bienhold U. Complexity of gap junctions between horizontal cells of the carp retina. Neuroscience 2016; 340:8-22. [PMID: 27793781 DOI: 10.1016/j.neuroscience.2016.10.044] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 10/11/2016] [Accepted: 10/18/2016] [Indexed: 11/17/2022]
Abstract
In the vertebrate retina, horizontal cells (HCs) reveal homologous coupling by gap junctions (gj), which are thought to consist of different connexins (Cx). However, recent studies in mouse, rabbit and zebrafish retina indicate that individual HCs express more than one connexin. To provide further insights into the composition of gj connecting HCs and to determine whether HCs express multiple connexins, we examined the molecular identity and distribution of gj between HCs of the carp retina. We have cloned four carp connexins designated Cx49.5, Cx55.5, Cx52.6 and Cx53.8 with a close relationship to connexins previously reported in HCs of mouse, rabbit and zebrafish, respectively. Using in situ hybridization, Cx49.5 expression was detected in different subpopulations of retinal neurons including HCs, whereas the Cx52.6 transcript was localized exclusively in HCs. Using specific antibodies, Cx55.5 and Cx53.8 were detected on dendrites of all four HC subtypes and axon terminals. Immunoelectron microscopy confirmed the presence of Cx55.5 and Cx53.8 in gap junctions between these processes and Cx55.5 was additionally observed in HC dendrites invaginating cone pedicles, suggesting its participation in the modulation of photoreceptor output in the carp retina. Furthermore, using single-cell RT-PCR, all four connexins were detected in different subtypes of HCs, suggesting overlapping expression patterns. Thus, the composition of gj mediating homologous coupling between subtypes of carp HCs appears to be more complex than expected. Moreover, BLAST searches of the preliminary carp genome, using novel sequences as query, suggest that most of the analyzed connexin genes are duplicated in carp.
Collapse
Affiliation(s)
- H Greb
- Visual Neuroscience, Department of Neuroscience, University of Oldenburg, D-26111 Oldenburg, Germany
| | - S Hermann
- Neurobiology, Department of Neuroscience, University of Oldenburg, D-26111 Oldenburg, Germany
| | - P Dirks
- Neurobiology, Department of Neuroscience, University of Oldenburg, D-26111 Oldenburg, Germany
| | - G Ommen
- Neurobiology, Department of Neuroscience, University of Oldenburg, D-26111 Oldenburg, Germany
| | - V Kretschmer
- Neurobiology, Department of Neuroscience, University of Oldenburg, D-26111 Oldenburg, Germany; Cell and Matrix Biology, Institute of Zoology, Johannes Gutenberg University of Mainz, D-55128 Mainz, Germany
| | - K Schultz
- Neurobiology, Department of Neuroscience, University of Oldenburg, D-26111 Oldenburg, Germany
| | - G Zoidl
- Department of Psychology, Faculty of Health, York University, Toronto, Canada
| | - R Weiler
- Neurobiology, Department of Neuroscience, University of Oldenburg, D-26111 Oldenburg, Germany; Research Center Neurosensory Science, University of Oldenburg, D-26111 Oldenburg, Germany
| | - U Janssen-Bienhold
- Visual Neuroscience, Department of Neuroscience, University of Oldenburg, D-26111 Oldenburg, Germany; Research Center Neurosensory Science, University of Oldenburg, D-26111 Oldenburg, Germany.
| |
Collapse
|
40
|
Wang L, Qiu YH, Zeng Y. Coding Properties of Three Intrinsically Distinct Retinal Ganglion Cells under Periodic Stimuli: A Computational Study. Front Comput Neurosci 2016; 10:102. [PMID: 27721751 PMCID: PMC5033956 DOI: 10.3389/fncom.2016.00102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 09/09/2016] [Indexed: 11/13/2022] Open
Abstract
As the sole output neurons in the retina, ganglion cells play significant roles in transforming visual information into spike trains, and then transmitting them to the higher visual centers. However, coding strategies that retinal ganglion cells (RGCs) adopt to accomplish these processes are not completely clear yet. To clarify these issues, we investigate the coding properties of three types of RGCs (repetitive spiking, tonic firing, and phasic firing) by two different measures (spike-rate and spike-latency). Model results show that for periodic stimuli, repetitive spiking RGC and tonic RGC exhibit similar spike-rate patterns. Their spike- rates decrease gradually with increased stimulus frequency, moreover, variation of stimulus amplitude would change the two RGCs' spike-rate patterns. For phasic RGC, it activates strongly at medium levels of frequency when the stimulus amplitude is low. While if high stimulus amplitude is applied, phasic RGC switches to respond strongly at low frequencies. These results suggest that stimulus amplitude is a prominent factor in regulating RGCs in encoding periodic signals. Similar conclusions can be drawn when analyzes spike-latency patterns of the three RGCs. More importantly, the above phenomena can be accurately reproduced by Hodgkin's three classes of neurons, indicating that RGCs can perform the typical three classes of firing dynamics, depending on the distinctions of ion channel densities. Consequently, model results from the three RGCs may be not specific, but can also applicable to neurons in other brain regions which exhibit part(s) or all of the Hodgkin's three excitabilities.
Collapse
Affiliation(s)
- Lei Wang
- Neuroscience and Intelligent Media Institute, Communication University of China Beijing, China
| | - Yi-Hong Qiu
- School of Biomedical Engineering, Shanghai Jiao Tong University Shanghai, China
| | - Yanjun Zeng
- Biomedical Engineering Center, Beijing University of Technology Beijing, China
| |
Collapse
|
41
|
Lane BJ, Samarth P, Ransdell JL, Nair SS, Schulz DJ. Synergistic plasticity of intrinsic conductance and electrical coupling restores synchrony in an intact motor network. eLife 2016; 5. [PMID: 27552052 PMCID: PMC5026470 DOI: 10.7554/elife.16879] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 08/22/2016] [Indexed: 01/12/2023] Open
Abstract
Motor neurons of the crustacean cardiac ganglion generate virtually identical, synchronized output despite the fact that each neuron uses distinct conductance magnitudes. As a result of this variability, manipulations that target ionic conductances have distinct effects on neurons within the same ganglion, disrupting synchronized motor neuron output that is necessary for proper cardiac function. We hypothesized that robustness in network output is accomplished via plasticity that counters such destabilizing influences. By blocking high-threshold K+ conductances in motor neurons within the ongoing cardiac network, we discovered that compensation both resynchronized the network and helped restore excitability. Using model findings to guide experimentation, we determined that compensatory increases of both GA and electrical coupling restored function in the network. This is one of the first direct demonstrations of the physiological regulation of coupling conductance in a compensatory context, and of synergistic plasticity across cell- and network-level mechanisms in the restoration of output. DOI:http://dx.doi.org/10.7554/eLife.16879.001 Neurons can communicate with each other by releasing chemicals called neurotransmitters, or by forming direct connections with each other known as gap junctions. These direct connections allow electrical impulses to flow from one neuron to another via pores in the membranes between the cells. Unlike communication via neurotransmitters, gap junctions are usually thought to be hard-wired and unchanging over the life of the animal. Lane et al. recorded electrical activity in a network of neurons that generates rhythmic heart contractions in the Jonah crab. Neurons in this network usually all fire an electrical impulse at the same time, which is crucial to make sure that the whole heart contracts at the same time. The experiments show that drugs that block potassium channel pores in the membrane cause the neurons to fire too much and at different times to each other. However, the network of neurons soon adapted to the changes caused by the drugs and returned to working as normal. Mimicking these changes in a computer model of the neuron network, together with experimental data, showed that changes to the gap junctions play a major role in restoring normal activity to the network. The next step following on from this research is to understand how a network of neurons ‘senses’ that it is not working normally and changes its electrical activity. DOI:http://dx.doi.org/10.7554/eLife.16879.002
Collapse
Affiliation(s)
- Brian J Lane
- Division of Biological Sciences, University of Missouri-Columbia, Columbia, United States
| | - Pranit Samarth
- Department of Electrical and Computer Engineering, University of Missouri-Columbia, Columbia, United States
| | - Joseph L Ransdell
- Division of Biological Sciences, University of Missouri-Columbia, Columbia, United States
| | - Satish S Nair
- Department of Electrical and Computer Engineering, University of Missouri-Columbia, Columbia, United States
| | - David J Schulz
- Division of Biological Sciences, University of Missouri-Columbia, Columbia, United States
| |
Collapse
|
42
|
Kántor O, Benkő Z, Énzsöly A, Dávid C, Naumann A, Nitschke R, Szabó A, Pálfi E, Orbán J, Nyitrai M, Németh J, Szél Á, Lukáts Á, Völgyi B. Characterization of connexin36 gap junctions in the human outer retina. Brain Struct Funct 2016; 221:2963-84. [PMID: 26173976 DOI: 10.1007/s00429-015-1082-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 07/06/2015] [Indexed: 10/23/2022]
Abstract
Retinal connexins (Cx) form gap junctions (GJ) in key circuits that transmit average or synchronize signals. Expression of Cx36, -45, -50 and -57 have been described in many species but there is still a disconcerting paucity of information regarding the Cx makeup of human retinal GJs. We used well-preserved human postmortem samples to characterize Cx36 GJ constituent circuits of the outer plexiform layer (OPL). Based on their location, morphometric characteristics and co-localizations with outer retinal neuronal markers, we distinguished four populations of Cx36 plaques in the human OPL. Three of these were comprised of loosely scattered Cx36 plaques; the distalmost population 1 formed cone-to-rod GJs, population 2 in the mid-OPL formed cone-to-cone GJs, whereas the proximalmost population 4 likely connected bipolar cell dendrites. The fourth population (population 3) of Cx36 plaques conglomerated beneath cone pedicles and connected dendritic tips of bipolar cells that shared a common presynaptic cone. Overall, we show that the human outer retina displays a diverse cohort of Cx36 GJ that follows the general mammalian scheme and display a great functional diversity.
Collapse
Affiliation(s)
- Orsolya Kántor
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Zsigmond Benkő
- Department of Theory, Wigner Research Center for Physics of the Hungarian Academy of Sciences, Budapest, 1121, Hungary
- Semmelweis University School of Ph.D. Studies, Budapest, 1085, Hungary
| | - Anna Énzsöly
- Department of Ophthalmology, Semmelweis University, Budapest, 1085, Hungary
- Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, 1094, Hungary
| | - Csaba Dávid
- Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, 1094, Hungary
| | - Angela Naumann
- Life Imaging Center, Center for Biological Systems Analysis, Albert-Ludwigs University, 79104, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, 79104, Freiburg, Germany
| | - Roland Nitschke
- Life Imaging Center, Center for Biological Systems Analysis, Albert-Ludwigs University, 79104, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, 79104, Freiburg, Germany
| | - Arnold Szabó
- Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, 1094, Hungary
| | - Emese Pálfi
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - József Orbán
- Department of Biophysics, University of Pécs, Pécs, 7624, Hungary
- János Szentágothai Research Center, University of Pécs, Ifjúság str. 6, 7624, Pécs, Hungary
| | - Miklós Nyitrai
- Department of Biophysics, University of Pécs, Pécs, 7624, Hungary
- János Szentágothai Research Center, University of Pécs, Ifjúság str. 6, 7624, Pécs, Hungary
| | - János Németh
- Department of Ophthalmology, Semmelweis University, Budapest, 1085, Hungary
| | - Ágoston Szél
- Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, 1094, Hungary
| | - Ákos Lukáts
- Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, 1094, Hungary
| | - Béla Völgyi
- János Szentágothai Research Center, University of Pécs, Ifjúság str. 6, 7624, Pécs, Hungary.
- MTA-PTE NAP B Retinal Electrical Synapses Research Group, Pécs, 7624, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, 7624, Hungary.
- Department of Ophthalmology, New York University Langone Medical Center, New York, NY, 10016, USA.
| |
Collapse
|
43
|
Koizumi A, Poznanski RR. Does heterogeneity of intracellular Ca[Formula: see text] dynamics underlie speed tuning of direction-selective responses in starburst amacrine cells? J Integr Neurosci 2016; 14:1-17. [PMID: 26762484 DOI: 10.1142/s0219635215500259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The starburst amacrine cell (SAC) plays a fundamental role in retinal motion perception. In the vertebrate retina, SAC dendrites have been shown to be directionally selective in terms of their Ca[Formula: see text] responses for stimuli that move centrifugally from the soma. The mechanism by which SACs show Ca[Formula: see text] bias for centrifugal motion is yet to be determined with precision. Recent morphological studies support a presynaptic delay in glutamate receptor activation induced Ca[Formula: see text] release from bipolar cells preferentially contacting SACs. However, bipolar cells are known to be electrotonically coupled so time delays between the bipolar cells that provide input to SACs seem unlikely. Using fluorescent microscopy and imunnostaining, we found that the endoplasmic reticulum (ER) is omnipresent in the soma extending to the distal processes of SACs. Consequently, a working hypothesis on heterogeneity of intracellular Ca[Formula: see text] dynamics from ER is proposed as a possible explanation for the cause of speed tuning of direction-selective Ca[Formula: see text] responses in dendrites of SACs.
Collapse
Affiliation(s)
- Amane Koizumi
- * National Institutes of Natural Sciences 105-0001, Tokyo, Japan
- † National Institute for Physiological Sciences Okazaki, Aichi 444-8585, Japan
| | - Roman R Poznanski
- ‡ Department of Clinical Sciences Faculty of Biosciences and Medical Engineering Universiti Teknologi Malaysia 81310 Johor Bahru, Malaysia
| |
Collapse
|
44
|
Decrock E, De Bock M, Wang N, Bultynck G, Giaume C, Naus CC, Green CR, Leybaert L. Connexin and pannexin signaling pathways, an architectural blueprint for CNS physiology and pathology? Cell Mol Life Sci 2015; 72:2823-51. [PMID: 26118660 PMCID: PMC11113968 DOI: 10.1007/s00018-015-1962-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 06/11/2015] [Indexed: 02/06/2023]
Abstract
The central nervous system (CNS) is composed of a highly heterogeneous population of cells. Dynamic interactions between different compartments (neuronal, glial, and vascular systems) drive CNS function and allow to integrate and process information as well as to respond accordingly. Communication within this functional unit, coined the neuro-glio-vascular unit (NGVU), typically relies on two main mechanisms: direct cell-cell coupling via gap junction channels (GJCs) and paracrine communication via the extracellular compartment, two routes to which channels composed of transmembrane connexin (Cx) or pannexin (Panx) proteins can contribute. Multiple isoforms of both protein families are present in the CNS and each CNS cell type is characterized by a unique Cx/Panx portfolio. Over the last two decades, research has uncovered a multilevel platform via which Cxs and Panxs can influence different cellular functions within a tissue: (1) Cx GJCs enable a direct cell-cell communication of small molecules, (2) Cx hemichannels and Panx channels can contribute to autocrine/paracrine signaling pathways, and (3) different structural domains of these proteins allow for channel-independent functions, such as cell-cell adhesion, interactions with the cytoskeleton, and the activation of intracellular signaling pathways. In this paper, we discuss current knowledge on their multifaceted contribution to brain development and to specific processes in the NGVU, including synaptic transmission and plasticity, glial signaling, vasomotor control, and blood-brain barrier integrity in the mature CNS. By highlighting both physiological and pathological conditions, it becomes evident that Cxs and Panxs can play a dual role in the CNS and that an accurate fine-tuning of each signaling mechanism is crucial for normal CNS physiology.
Collapse
Affiliation(s)
- Elke Decrock
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| | - Marijke De Bock
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| | - Nan Wang
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, KU Leuven, Louvain, Belgium
| | - Christian Giaume
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, 75231 Paris Cedex 05, France
- University Pierre et Marie
Curie, ED, N°158, 75005 Paris, France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, 75005 Paris, France
| | - Christian C. Naus
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Colin R. Green
- Department of Ophthalmology, The University of Auckland, Auckland, New Zealand
| | - Luc Leybaert
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| |
Collapse
|
45
|
Jin NG, Chuang AZ, Masson PJ, Ribelayga CP. Rod electrical coupling is controlled by a circadian clock and dopamine in mouse retina. J Physiol 2015; 593:1597-631. [PMID: 25616058 PMCID: PMC4386962 DOI: 10.1113/jphysiol.2014.284919] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 01/19/2015] [Indexed: 11/08/2022] Open
Abstract
Rod single-photon responses are critical for vision in dim light. Electrical coupling via gap junction channels shapes the light response properties of vertebrate photoreceptors, but the regulation of rod coupling and its impact on the single-photon response have remained unclear. To directly address these questions, we developed a perforated patch-clamp recording technique and recorded from single rod inner segments in isolated intact neural mouse retinae, maintained by superfusion. Experiments were conducted at different times of the day or under constant environmental conditions, at different times across the circadian cycle. We show that rod electrical coupling is regulated by a circadian clock and dopamine, so that coupling is weak during the day and strong at night. Altogether, patch-clamp recordings of single-photon responses in mouse rods, tracer coupling, receptive field measurements and pharmacological manipulations of gap junction and dopamine receptor activity provide compelling evidence that rod coupling is modulated in a circadian manner. These data are consistent with computer modelling. At night, single-photon responses are smaller due to coupling, but the signal-to-noise ratio for a dim (multiphoton) light response is increased at night because of signal averaging between coupled rods.
Collapse
Affiliation(s)
- Nan Ge Jin
- Ruiz Department of Ophthalmology and Visual Science, Medical School, The University of Texas Health Science Centre at Houston6431 Fannin Street, Suite MSB 7.024, Houston, TX, 77030, USA
| | - Alice Z Chuang
- Ruiz Department of Ophthalmology and Visual Science, Medical School, The University of Texas Health Science Centre at Houston6431 Fannin Street, Suite MSB 7.024, Houston, TX, 77030, USA
| | - Philippe J Masson
- Department of Mechanical Engineering, Cullen College of Engineering, University of HoustonN207 Engineering Building 1, Suite W204, Houston, TX, 77204, USA
| | - Christophe P Ribelayga
- Ruiz Department of Ophthalmology and Visual Science, Medical School, The University of Texas Health Science Centre at Houston6431 Fannin Street, Suite MSB 7.024, Houston, TX, 77030, USA
- Graduate School of Biomedical Sciences, The University of Texas Health Science Centre at Houston, 6767 Bertner Avenue, Mitchell BuildingBSRB Suite 3.8344, Houston, TX, 77030, USA
- Neuroscience Graduate Program, The University of Texas Health Science Centre at Houston, Medical School6431 Fannin Street, Suite MSB 7.262, Houston, TX, 77030, USA
- Neuroscience Research Centre, The University of Texas Health Science Centre at HoustonHouston, 6431 Fannin Street, Suite MSB 7.046, TX, 77030, USA
| |
Collapse
|
46
|
Jing Y, Guo S, Zhang X, Sun A, Tao F, Ju H, Qian H. Effects of small interfering RNA interference of connexin 37 on subcutaneous gastric tumours in mice. Mol Med Rep 2014; 10:2955-60. [PMID: 25310476 DOI: 10.3892/mmr.2014.2609] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 05/09/2014] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate the effects of small interfering (si)RNA interference of connexin 37 (Cx37) on subcutaneous gastric tumours in mice. Constructed lentiviruses carrying siRNA against Cx37 significantly knocked down Cx37 mRNA and protein expression in vitro. A total of 60 mice with gastric cancer were randomly divided into the Cx37 siRNA group, the mock‑siRNA group and the control group. Cx37 siRNA, mock‑siRNA and saline were separately injected (with the lentiviruses transfected into the gastric cancer cells). Following six weeks, the Cx37 mRNA expression, Cx37 protein expression and tumor apoptosis were detected using semiquantitative reverse transcription‑polymerase chain reaction, western blot analysis and terminal deoxynucleotidyl transferase‑mediated dUTP nick end labelling, respectively. Six weeks following lentiviral transfection, the Cx37 mRNA levels in the Cx37 siRNA group, mock‑siRNA group and saline group decreased to 42, 63 and 67%, respectively (P<0.05). The mock‑siRNA group demonstrated no significant change in Cx37 levels compared with the control group. Western blot analysis revealed lower Cx37 protein levels in the Cx37‑RNAi group than in the other groups (0.21±0.07 vs. 0.65±0.06 vs. 0.54±0.07), and that the apoptotic index of the Cx37‑RNAi group was higher than those of the mock‑siRNA and control groups (19.7±5.1 vs. 9.8±6.4 vs. 10.5±7.2%, 11.1±6.9; P<0.05). In conclusion, it was demonstrated that Cx37 siRNA is correlated with gastric cancer. Interference of Cx37 effectively reduces Cx37 mRNA and protein expression and promotes tumour apoptosis.
Collapse
Affiliation(s)
- Yuanming Jing
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, Zhejiang 312000, P.R. China
| | - Suxia Guo
- Department of Cardiology, The Affiliated People's Hospital of Nanjing Medical University in Wuxi and People's Hospital of Wuxi City, Wuxi, Jiangsu 214023, P.R. China
| | - Xiaoping Zhang
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, Zhejiang 312000, P.R. China
| | - Aijing Sun
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, Zhejiang 312000, P.R. China
| | - Feng Tao
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, Zhejiang 312000, P.R. China
| | - Haixing Ju
- Department of Colorectal Surgery, Zhejiang Provincial Tumor Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Haixin Qian
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
47
|
Kovács-Öller T, Raics K, Orbán J, Nyitrai M, Völgyi B. Developmental changes in the expression level of connexin36 in the rat retina. Cell Tissue Res 2014; 358:289-302. [PMID: 25110193 DOI: 10.1007/s00441-014-1967-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 07/09/2014] [Indexed: 02/03/2023]
Abstract
Connexin36 (Cx36) is the major gap junction forming protein in the brain and the retina; thus, alterations in its expression indicate changes in the corresponding circuitry. Many structural changes occur in the early postnatal retina before functional neuronal circuits are finalized, including those that incorporate gap junctions. To reveal the time-lapse formation of inner retinal gap junctions, we examine the developing postnatal rat retina from birth (P0) to young adult age (P20) and follow the expression of Cx36 in the mRNA and protein levels. We found a continuous elevation in the expression of both the Cx36 transcript and protein between P0 and P20 and a somewhat delayed Cx36 plaque formation throughout the inner plexiform layer (IPL) starting at P10. By using tristratificated calretinin positive (CaR(+)) fibers in the IPL as a guide, we detected a clear preference of Cx36 plaques for the ON sublamina from the earliest time of detection. This distributional preference became more pronounced at P15 and P20 due to the emergence and widespread expression of large (>0.1 μm(2)) Cx36 plaques in the ON sublamina. Finally, we showed that parvalbumin-positive (PV(+)) AII amacrine cell dendrites colocalize with Cx36 plaques as early as P10 in strata 3 and 4, whereas colocalizations in stratum 5 became characteristic only around P20. We conclude that Cx36 expression in the rat IPL displays a characteristic succession of changes during retinogenesis reflecting the formation of the underlying electrical synaptic circuitry. In particular, AII cell gap junctions, first formed with ON cone bipolar cells and later with other AII amacrine cells, accounted for the observed Cx36 expressional changes.
Collapse
Affiliation(s)
- Tamás Kovács-Öller
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Ifjúság street 6, Hungary
| | | | | | | | | |
Collapse
|
48
|
Kurtenbach S, Kurtenbach S, Zoidl G. Emerging functions of pannexin 1 in the eye. Front Cell Neurosci 2014; 8:263. [PMID: 25309318 PMCID: PMC4163987 DOI: 10.3389/fncel.2014.00263] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 08/14/2014] [Indexed: 01/23/2023] Open
Abstract
Pannexin 1 (Panx1) is a high-conductance, voltage-gated channel protein found in vertebrates. Panx1 is widely expressed in many organs and tissues, including sensory systems. In the eye, Panx1 is expressed in major divisions including the retina, lens and cornea. Panx1 is found in different neuronal and non-neuronal cell types. The channel is mechanosensitive and responds to changes in extracellular ATP, intracellular calcium, pH, or ROS/nitric oxide. Since Panx1 channels operate at the crossroad of major signaling pathways, physiological functions in important autocrine and paracrine feedback signaling mechanisms were hypothesized. This review starts with describing in depth the initial Panx1 expression and localization studies fostering functional studies that uncovered distinct roles in processing visual information in subsets of neurons in the rodent and fish retina. Panx1 is expressed along the entire anatomical axis from optical nerve to retina and cornea in glia, epithelial and endothelial cells as well as in neurons. The expression and diverse localizations throughout the eye points towards versatile functions of Panx1 in neuronal and non-neuronal cells, implicating Panx1 in the crosstalk between immune and neural cells, pressure related pathological conditions like glaucoma, wound repair or neuronal cell death caused by ischemia. Summarizing the literature on Panx1 in the eye highlights the diversity of emerging Panx1 channel functions in health and disease.
Collapse
Affiliation(s)
- Sarah Kurtenbach
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada
| | - Stefan Kurtenbach
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada
| | - Georg Zoidl
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada ; Department of Biology, Faculty of Science, York University Toronto, ON, Canada
| |
Collapse
|
49
|
Dunn FA, Wong ROL. Wiring patterns in the mouse retina: collecting evidence across the connectome, physiology and light microscopy. J Physiol 2014; 592:4809-23. [PMID: 25172948 DOI: 10.1113/jphysiol.2014.277228] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The visual system has often been thought of as a parallel processor because distinct regions of the brain process different features of visual information. However, increasing evidence for convergence and divergence of circuit connections, even at the level of the retina where visual information is first processed, chips away at a model of dedicated and distinct pathways for parallel information flow. Instead, our current understanding is that parallel channels may emerge, not from exclusive microcircuits for each channel, but from unique combinations of microcircuits. This review depicts diagrammatically the current knowledge and remaining puzzles about the retinal circuit with a focus on the mouse retina. Advances in techniques for labelling cells and genetic manipulations have popularized the use of transgenic mice. We summarize evidence gained from serial electron microscopy, electrophysiology and light microscopy to illustrate the wiring patterns in mouse retina. We emphasize the need to explore proposed retinal connectivity using multiple methods to verify circuits both structurally and functionally.
Collapse
Affiliation(s)
- Felice A Dunn
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, 94143-0730, USA
| | - Rachel O L Wong
- Department of Biological Structure, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
50
|
Palacios-Muñoz A, Escobar MJ, Vielma A, Araya J, Astudillo A, Valdivia G, García IE, Hurtado J, Schmachtenberg O, Martínez AD, Palacios AG. Role of connexin channels in the retinal light response of a diurnal rodent. Front Cell Neurosci 2014; 8:249. [PMID: 25202238 PMCID: PMC4142540 DOI: 10.3389/fncel.2014.00249] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 08/05/2014] [Indexed: 01/02/2023] Open
Abstract
Several studies have shown that connexin channels play an important role in retinal neural coding in nocturnal rodents. However, the contribution of these channels to signal processing in the retina of diurnal rodents remains unclear. To gain insight into this problem, we studied connexin expression and the contribution of connexin channels to the retinal light response in the diurnal rodent Octodon degus (degu) compared to rat, using in vivo ERG recording under scotopic and photopic light adaptation. Analysis of the degu genome showed that the common retinal connexins present a high degree of homology to orthologs expressed in other mammals, and expression of Cx36 and Cx43 was confirmed in degu retina. Cx36 localized mainly to the outer and inner plexiform layers (IPLs), while Cx43 was expressed mostly in cells of the retinal pigment epithelium. Under scotopic conditions, the b-wave response amplitude was strongly reduced by 18-β-glycyrrhetinic acid (β-GA) (−45.1% in degu, compared to −52.2% in rat), suggesting that connexins are modulating this response. Remarkably, under photopic adaptation, β-GA increased the ERG b-wave amplitude in degu (+107.2%) while reducing it in rat (−62.3%). Moreover, β-GA diminished the spontaneous action potential firing rate in ganglion cells (GCs) and increased the response latency of ON and OFF GCs. Our results support the notion that connexins exert a fine-tuning control of the retinal light response and have an important role in retinal neural coding.
Collapse
Affiliation(s)
- Angelina Palacios-Muñoz
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso Valparaíso, Chile
| | - Maria J Escobar
- Departamento de Electrónica, Universidad Técnico Federico Santa María Valparaíso, Chile
| | - Alex Vielma
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso Valparaíso, Chile
| | - Joaquín Araya
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso Valparaíso, Chile
| | - Aland Astudillo
- Departamento de Electrónica, Universidad Técnico Federico Santa María Valparaíso, Chile
| | - Gonzalo Valdivia
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso Valparaíso, Chile
| | - Isaac E García
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso Valparaíso, Chile
| | - José Hurtado
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso Valparaíso, Chile ; Instituto de Sistemas Complejos de Valparaíso Valparaíso, Chile
| | - Oliver Schmachtenberg
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso Valparaíso, Chile
| | - Agustín D Martínez
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso Valparaíso, Chile
| | - Adrian G Palacios
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso Valparaíso, Chile ; Instituto de Sistemas Complejos de Valparaíso Valparaíso, Chile
| |
Collapse
|